1
|
van Eijck CWF, Sabroso-Lasa S, Strijk GJ, Mustafa DAM, Fellah A, Koerkamp BG, Malats N, van Eijck CHJ. A liquid biomarker signature of inflammatory proteins accurately predicts early pancreatic cancer progression during FOLFIRINOX chemotherapy. Neoplasia 2024; 49:100975. [PMID: 38335839 PMCID: PMC10873733 DOI: 10.1016/j.neo.2024.100975] [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: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is often treated with FOLFIRINOX, a chemotherapy associated with high toxicity rates and variable efficacy. Therefore, it is crucial to identify patients at risk of early progression during treatment. This study aims to explore the potential of a multi-omics biomarker for predicting early PDAC progression by employing an in-depth mathematical modeling approach. METHODS Blood samples were collected from 58 PDAC patients undergoing FOLFIRINOX before and after the first cycle. These samples underwent gene (GEP) and inflammatory protein expression profiling (IPEP). We explored the predictive potential of exclusively IPEP through Stepwise (Backward) Multivariate Logistic Regression modeling. Additionally, we integrated GEP and IPEP using Bayesian Kernel Regression modeling, aiming to enhance predictive performance. Ultimately, the FOLFIRINOX IPEP (FFX-IPEP) signature was developed. RESULTS Our findings revealed that proteins exhibited superior predictive accuracy than genes. Consequently, the FFX-IPEP signature consisted of six proteins: AMN, BANK1, IL1RL2, ITGB6, MYO9B, and PRSS8. The signature effectively identified patients transitioning from disease control to progression early during FOLFIRINOX, achieving remarkable predictive accuracy with an AUC of 0.89 in an independent test set. Importantly, the FFX-IPEP signature outperformed the conventional CA19-9 tumor marker. CONCLUSIONS Our six-protein FFX-IPEP signature holds solid potential as a liquid biomarker for the early prediction of PDAC progression during toxic FOLFIRINOX chemotherapy. Further validation in an external cohort is crucial to confirm the utility of the FFX-IPEP signature. Future studies should expand to predict progression under different chemotherapies to enhance the guidance of personalized treatment selection in PDAC.
Collapse
Affiliation(s)
- Casper W F van Eijck
- Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Madrid, Spain.
| | - Sergio Sabroso-Lasa
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Madrid, Spain; Centro de Investigación Biomédica en Red-Cáncer (CIBERONC), Madrid, Spain
| | - Gaby J Strijk
- Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Dana A M Mustafa
- Department of Clinical Bioinformatics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Amine Fellah
- Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Madrid, Spain; Centro de Investigación Biomédica en Red-Cáncer (CIBERONC), Madrid, Spain
| | - Casper H J van Eijck
- Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Madrid, Spain.
| |
Collapse
|
2
|
Chen Z, Zhang J, Dong C, Li D, Yin Y, Yu W, Chen Y. TNFAIP8 regulates gastric cancer growth via mTOR-Akt-ULK1 pathway and autophagy signals. J Cell Mol Med 2021; 25:3361-3370. [PMID: 33682317 PMCID: PMC8034480 DOI: 10.1111/jcmm.16413] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/11/2021] [Accepted: 02/08/2021] [Indexed: 12/13/2022] Open
Abstract
In this study, the purpose of this study was to investigate the role of TNFAIP8 in gastric cancer (GC). The expression of TNFAIP8 was detected by RT-PCR or western blot . TNFAIP8 was silenced or overexpressed in two cell lines. CCK-8 assay, transwell assay and flow cytometry were used to analyse cell viability, cell invasion capability and apoptosis, respectively. Nude mice were inoculated with TNFAIP8 silencing or overexpressing cells to form transplanted tumours. HE staining and immunohistochemistry assay were performed to assess histopathological changes in tumours. We found that the mRNA and protein expression of TNFAIP8 were significantly up-regulated in GC tumour tissues and cells compared with the normal counterparts. Overexpression of TNFAIP8 in GC cells increased cell viability, decreased apoptosis and promoted the cell migration ability. Meanwhile, increased expression of TNFAIP8 promoted autophagy, while inhibiting mTOR-Akt-ULK1 signal pathway. In conclusions, this study presents data that TNFAIP8 inhibits GC cells presumably by down-regulating mTOR-Akt-ULK1 signal pathway and activating autophagy signal.
Collapse
Affiliation(s)
- Zheng Chen
- Tumor Research and Therapy CenterShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Jianguo Zhang
- Department of Gastrointestinal SurgeryLiaocheng Dongchangfu People's HospitalLiaochengChina
| | - Chenyang Dong
- Department of Gastrointestinal SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Dongsheng Li
- Department of Gastrointestinal SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Yuehan Yin
- Department of Gastrointestinal SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Wenhai Yu
- Department of Gastrointestinal SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Yuezhi Chen
- Department of Gastrointestinal SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| |
Collapse
|
3
|
Ma HY, Li Y, Yin HZ, Yin H, Qu YY, Xu QY. TNFAIP8 Promotes Cisplatin Chemoresistance in Triple-Negative Breast Cancer by Repressing p53-Mediated miR-205-5p Expression. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 22:640-656. [PMID: 33230463 PMCID: PMC7581818 DOI: 10.1016/j.omtn.2020.09.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/21/2020] [Indexed: 12/21/2022]
Abstract
Tumor necrosis factor alpha-induced protein 8 (TNFAIP8) is implicated in the tumor progression and prognosis of triple-negative breast cancer (TNBC), but the detailed regulatory mechanism of TNFAIP8 in cisplatin tolerance in TNBC has not yet been investigated. TNFAIP8 was evidently upregulated in TNBC tumor tissues and cell lines. Knocking down TNFAIP8 led to impaired proliferation and elevated apoptosis of TNBC cells upon cisplatin (DDP) treatment. Mechanistic studies revealed that TNFAIP8 repressed the expression of p53 and p53-promoted microRNA (miR)-205-5p; moreover, miR-205-5p targeted multiple genes required for the cell cycle and repressed Akt phosphorylation, which thus inhibited the proliferation of TNBC cells. In addition, silencing of TNFAIP8 led to the upregulation of miR-205-5p and the restraint of the TRAF2-NF-κB pathway, which thus enhanced the suppressive effects of DDP on tumor growth in nude mice. This study revealed that TNFAIP8 was essential in the DDP tolerance formation of TNBC cells by reducing p53-promoted miR-205-5p expression. Thus, targeting TNFAIP8 might become a promising strategy to suppress TNBC progression.
Collapse
Affiliation(s)
- Hong-Yu Ma
- Department of Breast Radiotherapy, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, P.R. China
| | - Yang Li
- Department of Breast Radiotherapy, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, P.R. China
| | - Hui-Zi Yin
- Department of Breast Radiotherapy, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, P.R. China
| | - Hang Yin
- Department of Breast Radiotherapy, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, P.R. China
| | - Yuan-Yuan Qu
- Department of Breast Radiotherapy, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, P.R. China
| | - Qing-Yong Xu
- Department of Breast Radiotherapy, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, P.R. China
| |
Collapse
|
4
|
Rastgoo N, Wu J, Liu A, Pourabdollah M, Atenafu EG, Reece D, Chen W, Chang H. Targeting CD47/TNFAIP8 by miR-155 overcomes drug resistance and inhibits tumor growth through induction of phagocytosis and apoptosis in multiple myeloma. Haematologica 2020; 105:2813-2823. [PMID: 33256380 PMCID: PMC7716364 DOI: 10.3324/haematol.2019.227579] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 11/27/2019] [Indexed: 12/04/2022] Open
Abstract
The mechanisms of drug resistance in multiple myeloma are poorly understood. Here we show that CD47, an integrin-associated receptor, is significantly upregulated in drug resistant myeloma cells in comparison with parental cells, and that high expression of CD47 detected by immunohistochemistry is associated with shorter progression free and overall survivals in multiple myeloma patients. We show that miR-155 is expressed at low levels in drug resistant myeloma cells and is a direct regulator of CD47 through its 3'UTR. Furthermore, low miR-155 levels are associated with advanced stages of disease. MiR-155 overexpression suppressed CD47 expression on myeloma cell surface, leading to induction of phagocytosis of myeloma cells by macrophages and inhibition of tumor growth. MiR-155 overexpression also re-sensitized drug-resistant myeloma cells to bortezomib leading to cell death through targeting TNFAIP8, a negative mediator of apoptosis in vitro and in vivo. Thus, miR-155 mimics may serve as a promising new therapeutic modality by promoting phagocytosis and inducing apoptosis in patients with refractory/relapsed multiple myeloma.
Collapse
Affiliation(s)
- Nasrin Rastgoo
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Jian Wu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Aijun Liu
- Department of Hematology, Beijing Chaoyang Hospital, Capital University Beijing, Beijing, China
| | - Maryam Pourabdollah
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Eshetu G. Atenafu
- Department of Biostatistics, University Health Network, Toronto, Ontario, Canada
| | - Donna Reece
- Department of Hematology and Medical Oncology, University Health Network, Toronto, Ontario, Canada
| | - Wenming Chen
- Department of Hematology, Beijing Chaoyang Hospital, Capital University Beijing, Beijing, China
| | - Hong Chang
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Department of Hematology, Beijing Chaoyang Hospital, Capital University Beijing, Beijing, China
| |
Collapse
|
5
|
Bordoloi D, Banik K, Vikkurthi R, Thakur KK, Padmavathi G, Sailo BL, Girisa S, Chinnathambi A, Alahmadi TA, Alharbi SA, Buhrmann C, Shakibaei M, Kunnumakkara AB. Inflection of Akt/mTOR/STAT-3 cascade in TNF-α induced protein 8 mediated human lung carcinogenesis. Life Sci 2020; 262:118475. [PMID: 32976884 DOI: 10.1016/j.lfs.2020.118475] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/13/2020] [Accepted: 09/17/2020] [Indexed: 02/08/2023]
Abstract
Lung cancer is the leading cause of cancer-related death across the globe. Despite the marked advances in detection and therapeutic approaches, management of lung cancer patients remains a major challenge to oncologists which can be mainly attributed to late stage diagnosis, tumor recurrence and chemoresistance. Therefore, to overthrow these limitations, there arises a vital need to develop effective biomarkers for the successful management of this aggressive cancer type. Notably, TNF-alpha induced protein 8 (TIPE), a nuclear factor-kappa B (NF-κB)-inducible, oncogenic molecule and cytoplasmic protein which is involved in the regulation of T lymphocyte-mediated immunity and different processes in tumor cells such as proliferation, cell death and evasion of growth suppressors, might serve as one such biomarker which would facilitate effective management of lung cancer. Expression studies revealed this protein to be significantly upregulated in different lung cancer types, pathological conditions, stages and grades of lung tumor compared to normal human lung tissues. In addition, knockout of TIPE led to the reduced proliferation, survival, invasion and migration of lung cancer cells. Furthermore, TIPE was found to function through modulation of Akt/mTOR/STAT-3 signaling cascade. This is the first report which shows the involvement of TIPE in tobacco induced lung carcinogenesis. It positively regulated nicotine, NNK, NNN, and BaP induced proliferation, survival and migration of lung cancer cells possibly via Akt/STAT-3 signaling. Thus, this protein possesses important role in the pathogenesis of lung tumor and hence it can be targeted for developing newer therapeutic interventions for the clinico-management of lung cancer.
Collapse
Affiliation(s)
- Devivasha Bordoloi
- Cancer Biology Laboratory, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India.
| | - Kishore Banik
- Cancer Biology Laboratory, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Rajesh Vikkurthi
- Cancer Biology Laboratory, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Krishan Kumar Thakur
- Cancer Biology Laboratory, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Ganesan Padmavathi
- Cancer Biology Laboratory, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Bethsebie Lalduhsaki Sailo
- Cancer Biology Laboratory, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Tahani Awad Alahmadi
- Department of Pediatrics, College of Medicine, King Saud University [Medical City], King Khalid University Hospital, PO Box-2925, Riyadh 11461, Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Constanze Buhrmann
- Department of Anatomy, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Mehdi Shakibaei
- Department of Anatomy, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India.
| |
Collapse
|
6
|
Gao H, Zhang Z, Jiang L, Zhang L, Qin L, Liu T, Yang S. TNFAIP8 variants as potential epidemiological and predictive biomarkers in ovarian cancer. Cancer Cell Int 2020; 20:396. [PMID: 32821249 PMCID: PMC7433149 DOI: 10.1186/s12935-020-01490-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 08/10/2020] [Indexed: 11/29/2022] Open
Abstract
Background This research aimed to investigate the association between tumor necrosis factor-a-induced protein 8 (TNFAIP8) polymorphisms and ovarian cancer (OC) susceptibility. Methods A case–control study of 210 patients with OC and 231 healthy controls was conducted to assess the association between TNFAIP8 polymorphisms (rs11064, rs1045241, and rs1045242) and OC risk in Heilongjiang Province of China. The SNaPshot SNP assay was conducted to detect SNP genotype. Logistic regression analysis was applied to illustrate the underlying association. Results Our research found that TNFAIP8 rs11064 and rs1045242 were significantly connected with the susceptibility of OC. Additionally, rs1045242 increased the risk of OC, while rs11064 performed a protective role in the risk of OC. Data revealed that rs1045242 strongly related with advanced FIGO stage, larger residual tumor, and the presence of recurrence. Conclusions TNFAIP8 genetic variants, which may play difference roles, were significantly associated with OC susceptibility. The underlying molecular mechanism needs be clarified with scientific evidence.
Collapse
Affiliation(s)
- Hongyu Gao
- Department of Gastroenterologic Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081 China
| | - Zhiran Zhang
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin Medical University, 150 Haping Road, Harbin, 150081 China
| | - Liangliang Jiang
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin Medical University, 150 Haping Road, Harbin, 150081 China
| | - Lei Zhang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081 China
| | - Ling Qin
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081 China
| | - Tianbo Liu
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin Medical University, 150 Haping Road, Harbin, 150081 China
| | - Shanshan Yang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, Harbin Medical University, 150 Haping Road, Harbin, 150081 China
| |
Collapse
|
7
|
TNFAIP8 influences the motor function in mice after spinal cord injury (SCI) through meditating inflammation dependent on AKT. Biochem Biophys Res Commun 2020; 528:234-241. [PMID: 32487318 DOI: 10.1016/j.bbrc.2020.05.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/21/2022]
Abstract
Spinal cord injury (SCI) is a devastating disease and causes tissue loss and neurologic dysfunction, contributing to high morbidity and disability among human. However, the underlying molecular mechanisms still remain unclear. Tumor necrosis factor-α-induced protein 8 (TNFAIP8) is a member of the TNFAIP8/TIPE family, and has been implicated in different diseases associated with inflammation, infection, and immunity. Nevertheless, its effects on SCI have not been well investigated. In our study, we found time course of TNFAIP8 following SCI in mice, along with time-dependent increases of pro-inflammatory cytokines. The in vitro results confirmed the up-regulation of TNFAIP8 induced by lipopolysaccharide (LPS). Subsequently, we found that reducing TNFAIP8 by transfection with its specific siRNA (siTNFAIP8) markedly alleviated cell viability and inflammatory response caused by LPS in mouse microglial BV2 cells. Importantly, LPS-enhanced activation of inhibitor of κBα/nuclear factor-κB (IκBα/NF-κB) and phosphoinositide 3-kinase/serine-threonine kinase (PI3K/AKT) signaling pathways was considerably blunted by siTNFAIP8. Intriguingly, our results further showed that siTNFAIP8-restrained inflammation and IκBα/NF-κB in LPS-stimulated BV2 cells were almost abolished by the pre-treatment of AKT activator SC-79, demonstrating that TNFAIP8-regulated inflammatory response was largely dependent on AKT activation. Then, the in vivo studies were performed using the wild type (WT) and TNFAIP8-knockout (KO) mice with or without SCI operation. Results showed that TNFAIP8-KO mice exhibited improved neuron injury and locomotor function along with decreased microglial activity. Furthermore, compared with the WT/SCI mice, the expression of pro-inflammatory cytokines in spinal cords was markedly down-regulated by TNFAIP8-deficiency through blocking IκBα/NF-κB and PI3K/AKT signaling pathways. Taken together, these findings elucidated the novel role of TNFAIP8 in regulating SCI via the AKT signaling, and thus TNFAIP8 may be served as a promising therapeutic target for SCI treatment.
Collapse
|
8
|
Guo F, Yuan Y. Tumor Necrosis Factor Alpha-Induced Proteins in Malignant Tumors: Progress and Prospects. Onco Targets Ther 2020; 13:3303-3318. [PMID: 32368089 PMCID: PMC7182456 DOI: 10.2147/ott.s241344] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/04/2020] [Indexed: 12/14/2022] Open
Abstract
Tumor necrosis factor (TNF) is the first cytokine used in tumor biotherapy, but TNF-related drugs are limited by the lack of specific targets. Tumor necrosis factor alpha-induced proteins (TNFAIPs), derived from TNF, is a protein family and participates in proliferation, invasion and metastasis of tumor cells. In order to better understand biological functions and potential roles of TNFAIPs in malignant tumors, this paper in the form of “Gene–Protein–Tumor correlation” summarizes the biological characteristics, physiological functions and mechanisms of TNFAIPs by searching National Center of Biotechnology Information, GeneCards, UniProt and STRING databases. The relationship between TNFAIPs and malignant tumors is analyzed, and protein–protein interaction diagram in members of TNFAIPs is drawn based on TNF for the first time. We find that TNF as a key factor is related to TNFAIP1, TNFAIP3, TNFAIP5, TNFAIP6, TNFAIP8 and TNFAIP9, which can be directly involved in activating TNFAIP1, TNFAIP5, TNFAIP8 and TNFAIP9. We confirm that the mechanism of TNFAIP1, TNFAIP2 and TNFAIP3 inducing tumors may be related to NF-κB signaling pathway, but the mechanism of tumor induction by other members of TNFAIPs is not clear. In the future, translational studies are needed to explore the mechanisms of TNF-TNFAIPs-tumors.
Collapse
Affiliation(s)
- Fang Guo
- Liaoning Provincial Education Department, Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, and Key Laboratory of Cancer Etiology and Prevention (China Medical University), Shenyang City, Liaoning Province, People's Republic of China.,Department of Oncology, PLA Cancer Center, General Hospital of Northern Theater Command, Shenyang City, Liaoning Province, People's Republic of China
| | - Yuan Yuan
- Liaoning Provincial Education Department, Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, and Key Laboratory of Cancer Etiology and Prevention (China Medical University), Shenyang City, Liaoning Province, People's Republic of China
| |
Collapse
|
9
|
Niture S, Gyamfi MA, Lin M, Chimeh U, Dong X, Zheng W, Moore J, Kumar D. TNFAIP8 regulates autophagy, cell steatosis, and promotes hepatocellular carcinoma cell proliferation. Cell Death Dis 2020; 11:178. [PMID: 32152268 PMCID: PMC7062894 DOI: 10.1038/s41419-020-2369-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 02/07/2023]
Abstract
Tumor necrosis factor-α-induced protein 8 (TNFAIP8) expression has been linked to tumor progression in various cancer types, but the detailed mechanisms of TNFAIP8 are not fully elucidated. Here we define the role of TNFAIP8 in early events associated with development of hepatocellular carcinoma (HCC). Increased TNFAIP8 levels in HCC cells enhanced cell survival by blocking apoptosis, rendering HCC cells more resistant to the anticancer drugs, sorafenib and regorafenib. TNFAIP8 also induced autophagy and steatosis in liver cancer cells. Consistent with these observations, TNFAIP8 blocked AKT/mTOR signaling and showed direct interaction with ATG3-ATG7 proteins. TNFAIP8 also exhibited binding with fatty acids and modulated expression of lipid/fatty-acid metabolizing enzymes. Chronic feeding of mice with alcohol increased hepatic levels of TNFAIP8, autophagy, and steatosis but not in high-fat-fed obese mice. Similarly, higher TNFAIP8 expression was associated with steatotic livers of human patients with a history of alcohol use but not in steatotic patients with no history of alcohol use. Our data indicate a novel role of TNFAIP8 in modulation of drug resistance, autophagy, and hepatic steatosis, all key early events in HCC progression.
Collapse
Affiliation(s)
- Suryakant Niture
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, Durham, NC, 27707, USA
| | - Maxwell A Gyamfi
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, Durham, NC, 27707, USA
| | - Minghui Lin
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, Durham, NC, 27707, USA
- Ningxia Medical University, Ningxia Hui Autonomous Region, Yinchuan, 750004, China
| | - Uchechukwu Chimeh
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, Durham, NC, 27707, USA
| | - Xialan Dong
- Department of Pharmaceutical Sciences, Bio-manufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University Durham, Durham, NC, 27707, USA
| | - Weifan Zheng
- Department of Pharmaceutical Sciences, Bio-manufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University Durham, Durham, NC, 27707, USA
| | - John Moore
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, Durham, NC, 27707, USA
| | - Deepak Kumar
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, Durham, NC, 27707, USA.
- Department of Pharmaceutical Sciences, North Carolina Central University Durham, Durham, NC, 27707, USA.
| |
Collapse
|
10
|
Zhong M, Li N, Qiu X, Ye Y, Chen H, Hua J, Yin P, Zhuang G. TIPE regulates VEGFR2 expression and promotes angiogenesis in colorectal cancer. Int J Biol Sci 2020; 16:272-283. [PMID: 31929755 PMCID: PMC6949158 DOI: 10.7150/ijbs.37906] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 10/13/2019] [Indexed: 12/14/2022] Open
Abstract
Background: Metastasis is the leading cause of death in colorectal cancer (CRC) patients. It is regulated mainly by tumor cell angiogenesis, and angiogenesis is caused by the binding of vascular endothelial growth factor (VEGF) to vascular endothelial growth factor receptor 2 (VEGFR2). Tumor necrosis factor-α-induced protein 8 (TNFAIP8, hereto after TIPE) plays an important role in tumorigenesis, development, and prognosis. However, the relationship between TIPE and VEGFR2 in CRC angiogenesis and the mechanism of action remain unknown. Method: In this study, we used quantitative real-time PCR, Western blotting and immunohistochemistry to detect TIPE and VEGFR2 expression in 55 specimens from CRC patients. We also used HCT116 CRC cells and human umbilical vein endothelial cells (HUVECs) for in vitro experiments by stably transducing shTIPE and shRNA control lentivirus into HCT116 cells, detecting VEGFR2 expression after TIPE knockdown and repurposing the culture supernatant as conditioned medium to stimulate angiogenesis of HUVECs. In vivo experiments with chicken chorioallantoic membranes (CAMs) and a nude mouse matrix subcutaneous tumor model were performed to validate the effects of TIPE on angiogenesis. Additionally, we analyzed the expression and phosphorylation levels of PDK1 and blocked PDK1 expression using inhibitors to determine whether TIPE-induced changes in VEGFR2-mediated angiogenesis acted via the PI3K-Akt pathway. Results: We found that TIPE and VEGFR2 are highly expressed in CRC and act as oncogenes. TIPE knockdown also downregulated VEGFR2 expression, which resulted in simultaneous inhibition of cell proliferation, cell migration and angiogenesis. Then, in vivo experiments further demonstrated that TIPE promotes angiogenesis in CRC. Finally, we found that TIPE promotes VEGFR2-mediated angiogenesis by upregulating PDK1 expression and phosphorylation and that blocking PDK1 expression can inhibit this process. Conclusion: TIPE promotes angiogenesis in CRC by regulating the expression of VEGFR2, which may be a target for antiangiogenic cancer therapy.
Collapse
Affiliation(s)
- Mengya Zhong
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Nini Li
- Department of Pathology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| | - Xingfeng Qiu
- Department of Gastrointestinal Surgery, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
| | - Yuhan Ye
- Department of Pathology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
| | - Huiyu Chen
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jianyu Hua
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Ping Yin
- Department of Pathology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
| | - Guohong Zhuang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China.,Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
11
|
Liu T, Jiang L, Yu L, Ge T, Wang J, Gao H. Association of TNFAIP8 gene polymorphisms with endometrial cancer in northern Chinese women. Cancer Cell Int 2019; 19:105. [PMID: 31043860 PMCID: PMC6480735 DOI: 10.1186/s12935-019-0827-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/13/2019] [Indexed: 12/31/2022] Open
Abstract
Background Tumor necrosis factor-a-induced protein 8 (TNFAIP8) presented a elevated expression in endometrial cancer (EC). However, the relationship of TNFAIP8 gene polymorphisms with EC risk remains unclear. This case–control study aimed to investigate the effect of single nucleotide polymorphisms (SNPs) in TNFAIP8 on northern Chinese women with EC. Methods SNP rs11064, rs1045241, and rs1045242 in TNFAIP8 were successfully genotyped in 248 cancer-free controls and 226 ECs by SNaPshot method, respectively. Logistic regression was performed to assess relationship of SNPs with EC risk. The relationships of SNPs with clinicopathological variables were evaluated by Chi-square test or Student’s t-test or Fisher’s text. Results The minor alleles of rs11064 in TNFAIP8 were strongly associated with EC risk, with adjust odds ratio (OR) of 1.719 (95% CI 1.180–2.506, P = 0.005). The minor allele of rs1045242 in the TNFAIP8 gene was strongly associated with with EC risk (adjust OR: 1.636, 95% CI 1.107–2.417, P = 0.014). rs11064 SNPs correlated with TNFAIP8 protein expression in EC (P = 0.015). For rs1045242, patients with AG + GG presented higher TNFAIP8 protein expression than that with AA (P = 0.020). It also showed that SNP rs11064 was associated with advanced FIGO stage (P = 0.001), deep myometrial invasion (P = 0.047), and lymph node metastasis (P = 0.048) under the codominant model in ECs. Conclusions SNP rs11064 in TNFAIP8 increased EC risk and significantly related with its protein expression in northern Chinese women. Electronic supplementary material The online version of this article (10.1186/s12935-019-0827-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tianbo Liu
- 1Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin Medical University, 150 Haping Road, Harbin, 150081 China
| | - Liangliang Jiang
- 1Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin Medical University, 150 Haping Road, Harbin, 150081 China
| | - Libo Yu
- 1Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin Medical University, 150 Haping Road, Harbin, 150081 China
| | - Tingting Ge
- 1Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin Medical University, 150 Haping Road, Harbin, 150081 China
| | - Jing Wang
- 1Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin Medical University, 150 Haping Road, Harbin, 150081 China
| | - Hongyu Gao
- 2Department of Gastroenterologic Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, 150 Haping Road, Harbin, 150081 China
| |
Collapse
|
12
|
Niture S, Dong X, Arthur E, Chimeh U, Niture SS, Zheng W, Kumar D. Oncogenic Role of Tumor Necrosis Factor α-Induced Protein 8 (TNFAIP8). Cells 2018; 8:cells8010009. [PMID: 30586922 PMCID: PMC6356598 DOI: 10.3390/cells8010009] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/19/2022] Open
Abstract
Tumor necrosis factor (TNF)-α-induced protein 8 (TNFAIP8) is a founding member of the TIPE family, which also includes TNFAIP8-like 1 (TIPE1), TNFAIP8-like 2 (TIPE2), and TNFAIP8-like 3 (TIPE3) proteins. Expression of TNFAIP8 is strongly associated with the development of various cancers including cancer of the prostate, liver, lung, breast, colon, esophagus, ovary, cervix, pancreas, and others. In human cancers, TNFAIP8 promotes cell proliferation, invasion, metastasis, drug resistance, autophagy, and tumorigenesis by inhibition of cell apoptosis. In order to better understand the molecular aspects, biological functions, and potential roles of TNFAIP8 in carcinogenesis, in this review, we focused on the expression, regulation, structural aspects, modifications/interactions, and oncogenic role of TNFAIP8 proteins in human cancers.
Collapse
Affiliation(s)
- Suryakant Niture
- Julius L. Chambers Biomedical Biotechnology Research Institute (BBRI), North Carolina Central University, Durham, NC 27707, USA.
| | - Xialan Dong
- Bio-manufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA.
| | - Elena Arthur
- Julius L. Chambers Biomedical Biotechnology Research Institute (BBRI), North Carolina Central University, Durham, NC 27707, USA.
| | - Uchechukwu Chimeh
- Julius L. Chambers Biomedical Biotechnology Research Institute (BBRI), North Carolina Central University, Durham, NC 27707, USA.
| | | | - Weifan Zheng
- Bio-manufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA.
| | - Deepak Kumar
- Julius L. Chambers Biomedical Biotechnology Research Institute (BBRI), North Carolina Central University, Durham, NC 27707, USA.
- Department of Pharmaceutical Sciences, North Carolina Central University, Durham, NC 27707, USA.
| |
Collapse
|
13
|
Wang J, Gao H, Liu G, Gu L, Yang C, Zhang F, Liu T. Tumor necrosis factor α–induced protein 8 expression as a predictor of prognosis and resistance in patients with advanced ovarian cancer treated with neoadjuvant chemotherapy. Hum Pathol 2018; 82:239-248. [DOI: 10.1016/j.humpath.2018.02.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 02/12/2018] [Accepted: 02/16/2018] [Indexed: 10/28/2022]
|
14
|
Xie Y, Zhou F, Zhao X. TNFAIP8 promotes cell growth by regulating the Hippo pathway in epithelial ovarian cancer. Exp Ther Med 2018; 16:4975-4982. [PMID: 30546405 PMCID: PMC6256973 DOI: 10.3892/etm.2018.6819] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 08/14/2018] [Indexed: 12/13/2022] Open
Abstract
Tumor necrosis factor-α-induced protein 8 (TNFAIP8) is an independent prognostic factor for cancer-specific and disease-free survival in patients with epithelial ovarian cancer (EOC). However, the exact mechanism of the biological role of TNFAIP8 in EOC remains unclear. In the present study, a siRNA specifically targeting TNFAIP8 was prepared to knock down TNFAIP8 in EOC cells. Cell growth, colony formation, apoptosis, and cell cycle distribution in TNFAIP8-deficient EOC cells were determined. In addition, the underlying molecular mechanisms were investigated by western blot analysis and reverse transcription quantitative polymerase chain reaction assays. It was demonstrated that the knockdown of TNFAIP8 inhibited EOC cell growth and colony formation, along with increased levels of apoptosis and cell cycle arrest. The results of the western blot analysis suggested that TNFAIP8 inhibited the expression of phosphorylated yes-associated protein 1 (YAP) while promoting total and nuclear YAP expression, followed by the regulation of apoptosis and cell cycle checkpoint protein expression in EOC. Overexpression of YAP in EOC cells efficiently attenuated cell growth inhibition in TNFAIP8-deficient EOC cells. In addition, knockdown of TNFAIP8 significantly impaired EOC tumor growth in vivo. Collectively, the data from the present study suggested that TNFAIP8 is an oncogene and a novel therapeutic target for EOC.
Collapse
Affiliation(s)
- Yao Xie
- Department of Gynecology and Obstetrics, Key Laboratory of Obstetrics and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China.,Department of Gynecology and Obstetrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Fei Zhou
- Department of Gynecology and Obstetrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Key Laboratory of Obstetrics and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
15
|
TIPE Family of Proteins and Its Implications in Different Chronic Diseases. Int J Mol Sci 2018; 19:ijms19102974. [PMID: 30274259 PMCID: PMC6213092 DOI: 10.3390/ijms19102974] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 09/19/2018] [Accepted: 09/22/2018] [Indexed: 12/14/2022] Open
Abstract
The tumor necrosis factor-α-induced protein 8-like (TIPE/TNFAIP8) family is a recently identified family of proteins that is strongly associated with the regulation of immunity and tumorigenesis. This family is comprised of four members, namely, tumor necrosis factor-α-induced protein 8 (TIPE/TNFAIP8), tumor necrosis factor-α-induced protein 8-like 1 (TIPE1/TNFAIP8L1), tumor necrosis factor-α-induced protein 8-like 2 (TIPE2/TNFAIP8L2), and tumor necrosis factor-α-induced protein 8-like 3 (TIPE3/TNFAIP8L3). Although the proteins of this family were initially described as regulators of tumorigenesis, inflammation, and cell death, they are also found to be involved in the regulation of autophagy and the transfer of lipid secondary messengers, besides contributing to immune function and homeostasis. Interestingly, despite the existence of a significant sequence homology among the four members of this family, they are involved in different biological activities and also exhibit remarkable variability of expression. Furthermore, this family of proteins is highly deregulated in different human cancers and various chronic diseases. This review summarizes the vivid role of the TIPE family of proteins and its association with various signaling cascades in diverse chronic diseases.
Collapse
|
16
|
Padmavathi G, Banik K, Monisha J, Bordoloi D, Shabnam B, Arfuso F, Sethi G, Fan L, Kunnumakkara AB. Novel tumor necrosis factor-α induced protein eight (TNFAIP8/TIPE) family: Functions and downstream targets involved in cancer progression. Cancer Lett 2018; 432:260-271. [DOI: 10.1016/j.canlet.2018.06.017] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 06/11/2018] [Accepted: 06/12/2018] [Indexed: 12/21/2022]
|
17
|
TNFAIP8 promotes the proliferation and cisplatin chemoresistance of non-small cell lung cancer through MDM2/p53 pathway. Cell Commun Signal 2018; 16:43. [PMID: 30064446 PMCID: PMC6069800 DOI: 10.1186/s12964-018-0254-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 07/19/2018] [Indexed: 12/29/2022] Open
Abstract
Background The highly refractory nature of non-small cell lung cancer (NSCLC) to chemotherapeutic drugs is an important factor resulting in its poor prognosis. Recent studies have revealed that tumour necrosis factor alpha-induced protein 8 (TNFAIP8) is involved in various biological and pathological processes of cells, but their underlying mechanisms in processes ranging from cancer development to drug resistance have not been fully elucidated. Methods TNFAIP8 expression in clinical NSCLC samples was examined through immunohistochemistry (IHC). After adjusting for patients’ characteristics with propensity score matching, Kaplan-Meier analysis and Cox regression analysis were performed for comparison of patients’ survival according to the TNFAIP8 level. Lentiviral transfection with TNFAIP8-specific shRNAs was used to establish stable TNFAIP8 knockdown (TNFAIP8 KD) NCI-H460, A549 and cis-diamminedichloroplatinum II resistant A549 (A549/cDDP) cell lines. Cell proliferation and viability were assessed by CCK-8 assay. Cell cycle was examined by flow cytometry. Multiple pathways regulated by TNFAIP8 KD were revealed by microarray analysis. Results We found that high TNFAIP8 expression was associated with advanced pT stage, advanced pTNM stage, lymph node metastasis and unfavourable survival in NSCLC patients. TNFAIP8 shRNAs reduced in vitro cancer cell proliferation and in vivo tumor growth. Additionally, TNFAIP8 KD increased the sensitivity of NSCLC cells to cisplatin in vitro and in vivo. Conversely, up-regulation of TNFAIP8 promoted the proliferation and drug resistance to cisplatin of NSCLC cells. TNFAIP8 influences cancer progression pathways involving the MDM2/p53 pathway. Indeed, we observed that TNFAIP8 KD mediated the MDM2 downregulation and the p53 ubiquitination, thereby decreasing the degradation of p53 protein. shRNA p53 reversed TNFAIP8 shRNA-mediated regulation of cell proliferation, cell cycle, cisplatin sensitivity, and expression levels of RAD51, a DNA repair gene. Conclusion Our work uncovers a hitherto unappreciated role of TNFAIP8 in NSCLC proliferation and cisplatin chemoresistance that is mediated through the MDM2/p53 pathway. These findings might offer potential therapeutic targets for reversing cisplatin resistance in NSCLC patients with high TNFAIP8 expression. Electronic supplementary material The online version of this article (10.1186/s12964-018-0254-x) contains supplementary material, which is available to authorized users.
Collapse
|
18
|
Niture S, Ramalinga M, Kedir H, Patacsil D, Niture SS, Li J, Mani H, Suy S, Collins S, Kumar D. TNFAIP8 promotes prostate cancer cell survival by inducing autophagy. Oncotarget 2018; 9:26884-26899. [PMID: 29928491 PMCID: PMC6003558 DOI: 10.18632/oncotarget.25529] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/03/2018] [Indexed: 01/11/2023] Open
Abstract
Tumor necrosis factor-α-inducible protein 8 (TNFAIP8) is a TNF-α inducible anti-apoptotic protein with multiple roles in tumor growth and survival. Mechanisms of cell survival by TNFAIP8 remain elusive. We investigated the role of TNFAIP8 in the regulation of the cell cycle, autophagy, cell survival and neuroendocrine differentiation in prostate cancer cells. We showed that TNFAIP8 dysregulates cell-cycle-related proteins, in PC3 cells. Oncogenic cell survival, drug resistance and dysregulation of cell cycle-related proteins are often associated with autophagy. We demonstrated that TNFAIP8 induces autophagy by increasing expression of autophagy effectors such as LC3β I/II, Beclin1, 4EBP1, p62, and SIRT1. We also demonstrated that TNFAIP8 interacts with autophagy-related protein 3 (ATG3). TNFα treatment increased the expression of TNFAIP8, which was associated with increased autophagy and decreased apoptosis. We also observed an increase in expression of neuroendocrine differentiation markers, synaptophysin and chromogranin A, and drug resistance to anticancer drugs, docetaxel and doxorubicin, in cells transfected with TNFAIP8. Collectively, our findings reveal that by the creation of cellular autophagy events, TNFAIP8 promotes cell survival and drug resistance in prostate cancer cells.
Collapse
Affiliation(s)
- Suryakant Niture
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, 27707 NC, USA.,Cancer Research Laboratory, University of the District of Columbia, Washington, 20008 DC, USA
| | - Malathi Ramalinga
- Cancer Research Laboratory, University of the District of Columbia, Washington, 20008 DC, USA
| | - Habib Kedir
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, 27707 NC, USA.,Cancer Research Laboratory, University of the District of Columbia, Washington, 20008 DC, USA
| | - Dorrelyn Patacsil
- Cancer Research Laboratory, University of the District of Columbia, Washington, 20008 DC, USA
| | | | - James Li
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, 20008 DC, USA
| | - Haresh Mani
- Department of Pathology, Inova Fairfax Hospital, Falls Church, 22042 VA, USA
| | - Simeng Suy
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, 20008 DC, USA
| | - Sean Collins
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, 20008 DC, USA
| | - Deepak Kumar
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, 27707 NC, USA.,Cancer Research Laboratory, University of the District of Columbia, Washington, 20008 DC, USA.,Lombardi Comprehensive Cancer Center, Georgetown University, Washington, 20008 DC, USA
| |
Collapse
|
19
|
Zhang L, Liu R, Luan YY, Yao YM. Tumor Necrosis Factor-α Induced Protein 8: Pathophysiology, Clinical Significance, and Regulatory Mechanism. Int J Biol Sci 2018; 14:398-405. [PMID: 29725261 PMCID: PMC5930472 DOI: 10.7150/ijbs.23268] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 02/26/2018] [Indexed: 12/13/2022] Open
Abstract
Tumor necrosis factor-α-induced protein-8 (TNFAIP8) is the earliest discovered component of TNFAIP8 family [tumor necrosis factor-α-induced protein-8 like (TIPE) family]. TNFAIP8 contains a putative death effector domain (DED) homologous to DED II in FLIP (Fas-associated death domain-like interleukin-1β-converting enzyme-inhibitory protein), which may affect cell survival/death process. Recently, it has been demonstrated that TNFAIP8 could inhibit apoptosis and autophagy in various types of cells. Moreover, TNFAIP8 level fluctuated evidently in patients with inflammatory, malignant, and autoimmune diseases, indicating that it might be an anti-apoptotic and oncogenetic protein. Herein we will review the discovery, gene/protein structure, pathophysiological functions, and clinical significance of TNFAIP8 together with its potential regulatory mechanism.
Collapse
Affiliation(s)
- Lei Zhang
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing 100048, People's Republic of China.,Emergency Department, The General Hospital of the Chinese PLA Rocket Force, Beijing 100088, People's Republic of China
| | - Ran Liu
- Department of Endocrinology, 307th Hospital of the Chinese PLA, Beijing 100071, People's Republic of China
| | - Ying-Yi Luan
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Yong-Ming Yao
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| |
Collapse
|
20
|
Wu X, Ma Y, Cheng J, Li X, Zheng H, Jiang L, Zhou R. TIPE1 function as a prognosis predictor and negative regulator of lung cancer. Oncotarget 2017; 8:78496-78506. [PMID: 29108244 PMCID: PMC5667977 DOI: 10.18632/oncotarget.19655] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 05/23/2017] [Indexed: 12/01/2022] Open
Abstract
TIPE1 (tumor necrosis factor-α-induced protein 8-like 1 or TNFAIP8L1) belongs to the TIPE (TNFAIP8) family, which act as a regulator of cell death. However, the expression and biologic functions of TIPE1 in lung cancer are largely unknown. Here, we investigated the roles of TIPE1 in lung cancer. Evaluated by qRT-PCR and immunohistochemical staining, lower TIPE1 mRNA and protein expression was found in the lung tumor tissue, compared with adjacent non-tumor tissues, which positively correlated with tumor patient survival. Overexpression of TIPE1 by lentivirus system in TIPE1-downregulated lung cancer cells significantly diminished cell growth and colony formation, companied with proliferation inhibition, apoptosis induction and invasion inhibition. It was identified to be due to TIPE1-regulated Cyclin D1, Cyclin B1, caspase 8, Caspase3, MM2 and MMP9 expression. Consistently, using a homograft tumor model in Balb/c mice, we discovered that TIPE1 prevented the growth and tumor weight of murine lung cancer homografts. Our findings revealed the anti-tumor role of TIPE1 in lung cancer cells and TIPE1 might be a novel prognostic indicator for lung cancer patients.
Collapse
Affiliation(s)
- Xiaocheng Wu
- Department of Emergency, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Yunmiao Ma
- Departmnet of Orthopedics, Zhuji People's Hospital, Zhejiang 311800, China
| | - Ji Cheng
- Department of Emergency, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Xia Li
- Department of Emergency, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Hui Zheng
- Department of Emergency, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Li Jiang
- Department of Emergency, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Renjie Zhou
- Department of Emergency, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| |
Collapse
|
21
|
Xiao M, Xu Q, Lou C, Qin Y, Ning X, Liu T, Zhao X, Jia S, Huang Y. Overexpression of TNFAIP8 is associated with tumor aggressiveness and poor prognosis in patients with invasive ductal breast carcinoma. Hum Pathol 2017; 62:40-49. [DOI: 10.1016/j.humpath.2016.12.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 12/21/2016] [Accepted: 12/30/2016] [Indexed: 02/04/2023]
|
22
|
Gao HY, Huo FC, Wang HY, Pei DS. MicroRNA-9 inhibits the gastric cancer cell proliferation by targeting TNFAIP8. Cell Prolif 2017; 50. [PMID: 28127811 DOI: 10.1111/cpr.12331] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 12/16/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND OBJECTIVES MicroRNA-9 is frequently dysregulated in many human carcinoma types, including gastric cancer (GC). Previous studies demonstrated that the expression of TNFAIP8 in GC is correlated with tumour occurrence, development, invasion, metastasis and prognosis. However, till now, the relationship between MicroRNA-9 and TNFAIP8 in GC has not been reported. MATERIALS AND METHODS Levels of miR-9 and TNFAIP8 expression in GC tissues and in human GC cell lines were studied using qualitative real-time PCR (qRT-PCR) and Western blotting. Cell viability was detected using the CCK-8 and clone formation assays. A dual-luciferase reporter system was used to confirm the target gene of miR-9. RESULTS We found that the expression level of MicroRNA-9 in GC tissues and cell lines was significantly lower than that in adjacent non-cancerous tissues and human immortalized gastric epithelial cell (GES) line, respectively. In addition, overexpression of MicroRNA-9 markedly inhibited GC cell proliferation in vitro and tumour growth in vivo. Further experiments revealed that TNFAIP8 was a direct and functional target of MicroRNA-9 in GC and overexpression of MicroRNA-9 obviously down-regulated the expression of TNFAIP8, which was involved in the gastric carcinogenesis and cancer progression. CONCLUSION Our results suggested that MicroRNA-9-TNFAIP8 might represent a promising diagnostic biomarker for GC patients and could be a potential therapeutic target in the prevention and treatment of GC.
Collapse
Affiliation(s)
- Hong-Yu Gao
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical University, Xuzhou, China
| | - Fu-Chun Huo
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical University, Xuzhou, China
| | - Hai-Yan Wang
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Dong-Sheng Pei
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical University, Xuzhou, China.,Department of Pathology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
23
|
Lowe JM, Nguyen TA, Grimm SA, Gabor KA, Peddada SD, Li L, Anderson CW, Resnick MA, Menendez D, Fessler MB. The novel p53 target TNFAIP8 variant 2 is increased in cancer and offsets p53-dependent tumor suppression. Cell Death Differ 2016; 24:181-191. [PMID: 27834950 DOI: 10.1038/cdd.2016.130] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 09/02/2016] [Accepted: 10/11/2016] [Indexed: 02/07/2023] Open
Abstract
Tumor necrosis factor-α-induced protein 8 (TNFAIP8) is a stress-response gene that has been associated with cancer, but no studies have differentiated among or defined the regulation or function of any of its several recently described expression variants. We found that TNFAIP8 variant 2 (v2) is overexpressed in multiple human cancers, whereas other variants are commonly downregulated in cancer (v1) or minimally expressed in cancer or normal tissue (v3-v6). Silencing v2 in cancer cells induces p53-independent inhibition of DNA synthesis, widespread binding of p53, and induction of target genes and p53-dependent cell cycle arrest and DNA damage sensitization. Cell cycle arrest induced by v2 silencing requires p53-dependent induction of p21. In response to the chemotherapeutic agent doxorubicin, p53 regulates v2 through binding to an intragenic enhancer, together indicating that p53 and v2 engage in complex reciprocal regulation. We propose that TNFAIP8 v2 promotes human cancer by broadly repressing p53 function, in essence offsetting p53-dependent tumor suppression.
Collapse
Affiliation(s)
- Julie M Lowe
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Thuy-Ai Nguyen
- Genome Integrity & Structural Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Sara A Grimm
- Biostatistics and Computational Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Kristin A Gabor
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Shyamal D Peddada
- Biostatistics and Computational Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Leping Li
- Biostatistics and Computational Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Carl W Anderson
- Genome Integrity & Structural Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Michael A Resnick
- Genome Integrity & Structural Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Daniel Menendez
- Genome Integrity & Structural Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Michael B Fessler
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| |
Collapse
|
24
|
Monteith JA, Mellert H, Sammons MA, Kuswanto LA, Sykes SM, Resnick-Silverman L, Manfredi JJ, Berger SL, McMahon SB. A rare DNA contact mutation in cancer confers p53 gain-of-function and tumor cell survival via TNFAIP8 induction. Mol Oncol 2016; 10:1207-20. [PMID: 27341992 DOI: 10.1016/j.molonc.2016.05.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 05/23/2016] [Accepted: 05/24/2016] [Indexed: 01/15/2023] Open
Abstract
The p53 tumor suppressor gene encodes a sequence-specific transcription factor. Mutations in the coding sequence of p53 occur frequently in human cancer and often result in single amino acid substitutions (missense mutations) in the DNA binding domain (DBD), blocking normal tumor suppressive functions. In addition to the loss of canonical functions, some missense mutations in p53 confer gain-of-function (GOF) activities to tumor cells. While many missense mutations in p53 cluster at six "hotspot" amino acids, the majority of mutations in human cancer occur elsewhere in the DBD and at a much lower frequency. We report here that mutations at K120, a non-hotspot DNA contact residue, confer p53 with the previously unrecognized ability to bind and activate the transcription of the pro-survival TNFAIP8 gene. Mutant K120 p53 binds the TNFAIP8 locus at a cryptic p53 response element that is not occupied by wild-type p53. Furthermore, induction of TNFAIP8 is critical for the evasion of apoptosis by tumor cells expressing the K120R variant of p53. These findings identify induction of pro-survival targets as a mechanism of gain-of-function activity for mutant p53 and will likely broaden our understanding of this phenomenon beyond the limited number of GOF activities currently reported for hotspot mutants.
Collapse
Affiliation(s)
- Jessica A Monteith
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, 233 S 10th Street, Philadelphia, PA 19107, United States.
| | - Hestia Mellert
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, 233 S 10th Street, Philadelphia, PA 19107, United States.
| | - Morgan A Sammons
- Cell and Developmental Biology, Epigenetics Program, Perelman School of Medicine, University of Pennsylvania, 9-125 Smilow Center for Translational Research, Philadelphia, PA 19104, United States.
| | - Laudita A Kuswanto
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, 233 S 10th Street, Philadelphia, PA 19107, United States; University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, United States.
| | - Stephen M Sykes
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, 233 S 10th Street, Philadelphia, PA 19107, United States; Medical Genetics and Molecular Biology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, United States.
| | - Lois Resnick-Silverman
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - James J Manfredi
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States.
| | - Shelley L Berger
- Cell and Developmental Biology, Epigenetics Program, Perelman School of Medicine, University of Pennsylvania, 9-125 Smilow Center for Translational Research, Philadelphia, PA 19104, United States.
| | - Steven B McMahon
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, 233 S 10th Street, Philadelphia, PA 19107, United States.
| |
Collapse
|
25
|
Shen P, Zhang H, Su Z, Wang S, Xu H. In Silico Analysis of Tumor Necrosis Factor α-Induced Protein 8-Like-1 (TIPE1) Protein. PLoS One 2015. [PMID: 26207809 PMCID: PMC4514785 DOI: 10.1371/journal.pone.0134114] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Tumor necrosis factor α-induced protein 8 (TNFAIP8)-like protein 1 (TIPE1) was a member of TNFAIP8 family. Previous studies have shown that TIPE1 could induce apoptosis in hepatocellular carcinoma. In this study, we attempted to predict its potential structure. Bioinformatic analysis of TIPE1 was performed to predict its potential structure using the bioinfomatic web services or softwares. The results showed that the amino acid sequences of TIPE1 were well conserved in mammals. No signal peptide and no transmembrane domain existed in human TIPE1. The aliphatic index of TIPE1 was 100.75 and the theoretical pI was 9.57. TIPE1 was a kind of stable protein and its grand average of hydropathicity was -0.108. Various post-translational modifications were also speculated to exist in TIPE1. In addition, the results of Swiss-Model Server and Swiss-Pdb Viewer program revealed that the predicted three-dimensional structure of TIPE1 protein was stable and it may accord with the rule of stereochemistry. TIPE1 was predicted to interact with FBXW5, caspase8 and so on. In conclusion, TIPE1 may be a stable protein with no signal peptide and no transmembrane domain. The bioinformatic analysis of TIPE1 will provide the basis for the further study on the function of TIPE1.
Collapse
Affiliation(s)
- Pei Shen
- Department of Immunology, Institute of Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu, People’s Republic of China
| | - Hong Zhang
- Department of Clinical Laboratory, Nantong Rich Hospital, The Fourth Clinical College of Yangzhou University, Nantong, Jiangsu, People’s Republic of China
| | - Zhaoliang Su
- Department of Immunology, Institute of Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Shengjun Wang
- Department of Immunology, Institute of Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Huaxi Xu
- Department of Immunology, Institute of Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
- * E-mail: (Huaxi Xu)
| |
Collapse
|
26
|
Cheng Y, Yu P, Duan X, Liu C, Xu S, Chen Y, Tan Y, Qiang Y, Shen J, Tao Z. Genome-wide analysis of androgen receptor binding sites in prostate cancer cells. Exp Ther Med 2015; 9:2319-2324. [PMID: 26136980 DOI: 10.3892/etm.2015.2406] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 03/20/2015] [Indexed: 01/23/2023] Open
Abstract
The transformation of prostate cancer from an androgen-dependent state to an androgen-independent state is a lethal progression. Alterations in transcriptional programs are the basis of prostate cancer deterioration. The androgen receptor (AR), a member of the nuclear hormone receptor superfamily, mediates prostate cancer progression by functioning primarily through the ligand-activated transcription of target genes. Therefore, a detailed map of AR-regulated genes and AR genomic binding sites is required for hormone-naive and castration-resistant prostate cancer. Through the use of chromatin immunoprecipitation in combination with direct sequencing, 4,143 AR binding sites were defined in the LNCaP androgen-sensitive prostate cancer cell line. Using the same method, 2,380 AR binding regions were identified in the LNCaP-AI long-term androgen-deprived cell line. Approximately 8.5% (354/4,143) of the binding regions were mapped to within 2 kb of the transcription start site (TSS) in the LNCaP cells, while ∼12.6% (299/2,380) were mapped to within 2 kb of the TSS in the LNCaP-AI cells. In total, the study mapped 2,796 genes in LNCaP cells and 1,854 genes in LNCaP-AI cells. The cell lines shared 789 mutual genes. In addition, gene ontology (GO) analysis of the genes revealed that there was a notable overlap between the GO terms in the LNCaP cells and LNCaP-AI cells. However, GO terms within the biological process domain that were only observed in the LNCaP-AI cells included the reproduction process, death, immune system process, multi-organism process, pigmentation and viral reproduction. The major genes in the different GO terms were TNFAIP8, RTN4, APP and SYNE1. Through analyzing the AR binding sites in the two cell types, the present study aimed to map potential AR-regulated genes, identify their associated transcription factors and provide a new perspective on the biological processes in the development of prostate cancer. The results provided a valuable data set that furthered the understanding of the genome-wide analysis of AR binding sites in prostate cancer cells, which may be exploited for the development of novel prostate cancer therapeutic strategies.
Collapse
Affiliation(s)
- Yue Cheng
- Department of Laboratory Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Pan Yu
- Department of Laboratory Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Xiuzhi Duan
- Department of Laboratory Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Chunhua Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Siqi Xu
- Department of Laboratory Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Yuhua Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Yunnian Tan
- Department of Laboratory Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Yun Qiang
- Department of Laboratory Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Junfang Shen
- Department of Laboratory Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Zhihua Tao
- Department of Laboratory Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
27
|
Bustin S, Dhillon HS, Kirvell S, Greenwood C, Parker M, Shipley GL, Nolan T. Variability of the reverse transcription step: practical implications. Clin Chem 2014; 61:202-12. [PMID: 25361949 DOI: 10.1373/clinchem.2014.230615] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND The reverse transcription (RT) of RNA to cDNA is a necessary first step for numerous research and molecular diagnostic applications. Although RT efficiency is known to be variable, little attention has been paid to the practical implications of that variability. METHODS We investigated the reproducibility of the RT step with commercial reverse transcriptases and RNA samples of variable quality and concentration. We quantified several mRNA targets with either singleplex SYBR Green I or dualplex probe-based reverse transcription real-time quantitative PCR (RT-qPCR), with the latter used to calculate the correlation between quantification cycles (Cqs) of mRNA targets amplified in the same real-time quantitative PCR (qPCR) assay. RESULTS RT efficiency is enzyme, sample, RNA concentration, and assay dependent and can lead to variable correlation between mRNAs from the same sample. This translates into relative mRNA expression levels that generally vary between 2- and 3-fold, although higher levels are also observed. CONCLUSIONS Our study demonstrates that the variability of the RT step is sufficiently large to call into question the validity of many published data that rely on quantification of cDNA. Variability can be minimized by choosing an appropriate RTase and high concentrations of RNA and characterizing the variability of individual assays by use of multiple RT replicates.
Collapse
Affiliation(s)
- Stephen Bustin
- Postgraduate Medical Institute, Faculty of Medical Science, Anglia Ruskin University, Chelmsford, UK;
| | - Harvinder S Dhillon
- Postgraduate Medical Institute, Faculty of Medical Science, Anglia Ruskin University, Chelmsford, UK
| | - Sara Kirvell
- Postgraduate Medical Institute, Faculty of Medical Science, Anglia Ruskin University, Chelmsford, UK
| | - Christina Greenwood
- Postgraduate Medical Institute, Faculty of Medical Science, Anglia Ruskin University, Chelmsford, UK
| | - Michael Parker
- Postgraduate Medical Institute, Faculty of Medical Science, Anglia Ruskin University, Chelmsford, UK
| | | | - Tania Nolan
- Institute of Population Health, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK
| |
Collapse
|