1
|
Zhao D, Song J, Ji C. Endoplasmic reticulum stress regulates apoptosis and chemotherapeutic via enhancing TNFRSF10B recycling to the cell membrane in triple-negative breast cancer. Clin Transl Oncol 2025; 27:265-276. [PMID: 38967737 DOI: 10.1007/s12094-024-03509-1] [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: 01/23/2024] [Accepted: 04/26/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is the most common malignant tumor in China. The expression and cell surface levels of TNF receptor superfamily member 10B (TNFRSF10B) are associated with apoptosis and chemotherapy. However, the precise molecular mechanisms that govern the regulation of TNFRSF10B remain unclear. MATERIALS AND METHODS RNA-Seq data related to TNBC chemotherapy resistance were acquired from the GEO database. The mRNA and protein levels of TNFRSF10B were detected using RT-PCR and Western blotting, respectively. Cell Counting Kit-8 (CCK-8) and colony formation assays were used to detect cell proliferation. Annexin V/7-AAD staining was used to evaluate apoptosis. The cell membrane TNFRSF10B was analyzed by Western blotting and immunofluorescence. Inducers and inhibitors of endoplasmic reticulum stress (ERS) were used to assess the effect of ERS on TNFRSF10B localization. RESULTS TNFRSF10B expression was downregulated in TNBC and was associated with prognosis. TNFRSF10B overexpression inhibits the growth of TNBC both in vivo and in vitro and can partially counteract chemotherapy resistance. ERS activation in TNBC promotes the expression of TNFRSF10B, leading to its enrichment on the cell membrane surface, thereby activating the apoptotic pathways. CONCLUSION ERS regulates the expression and subcellular localization of TNFRSF10B in TNBC cells. They synergistically affect anti-apoptosis and chemotherapy resistance in TNBC cells.
Collapse
Affiliation(s)
- Dapeng Zhao
- Breast Surgery (4Th General) Unit, General Surgery Department, General Hospital of Fushun Mining Bureau of Liaoning Health Industry, No.24 Centre Street, Xinfu District, Fushun City, 113008, Liaoning Province, China.
| | - Jian Song
- Breast Surgery (4Th General) Unit, General Surgery Department, General Hospital of Fushun Mining Bureau of Liaoning Health Industry, No.24 Centre Street, Xinfu District, Fushun City, 113008, Liaoning Province, China
| | - Chongyao Ji
- Breast Surgery (4Th General) Unit, General Surgery Department, General Hospital of Fushun Mining Bureau of Liaoning Health Industry, No.24 Centre Street, Xinfu District, Fushun City, 113008, Liaoning Province, China
| |
Collapse
|
2
|
Artykov AA, Yagolovich AV, Dolgikh DA, Kirpichnikov MP, Trushina DB, Gasparian ME. Death Receptors DR4 and DR5 Undergo Spontaneous and Ligand-Mediated Endocytosis and Recycling Regardless of the Sensitivity of Cancer Cells to TRAIL. Front Cell Dev Biol 2021; 9:733688. [PMID: 34660590 PMCID: PMC8514705 DOI: 10.3389/fcell.2021.733688] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/06/2021] [Indexed: 11/29/2022] Open
Abstract
Tumor necrosis factor-associated ligand inducing apoptosis (TRAIL) induces apoptosis through the death receptors (DRs) 4 and 5 expressed on the cell surface. Upon ligand stimulation, death receptors are rapidly internalized through clathrin-dependent and -independent mechanisms. However, there have been conflicting data on the role of death receptor endocytosis in apoptotic TRAIL signaling and possible cell type-specific differences in TRAIL signaling have been proposed. Here we have compared the kinetics of TRAIL-mediated internalization and subsequent recycling of DR4 and DR5 in resistant (HT-29 and A549) and sensitive (HCT116 and Jurkat) tumor cell lines of various origin. TRAIL stimulated the internalization of both receptors in a concentration-dependent manner with similar kinetics in sensitive and resistant cell lines without affecting the steady-state expression of DR4 and DR5 in cell lysates. Using the receptor-selective TRAIL variant DR5-B, we have shown that DR5 is internalized independently of DR4 receptor. After internalization and elimination of TRAIL from culture medium, the receptors slowly return to the plasma membrane. Within 4 h in resistant or 6 h in sensitive cells, the surface expression of receptors was completely restored. Recovery of receptors occurred both from newly synthesized molecules or from trans-Golgi network, as cycloheximide and brefeldin A inhibited this process. These agents also suppressed the expression of cell surface receptors in a time- and concentration-dependent manner, indicating that DRs undergo constitutive endocytosis. Inhibition of receptor endocytosis by sucrose led to sensitization of resistant cells to TRAIL and to an increase in its cytotoxic activity against sensitive cells. Our results confirm the universal nature of TRAIL-induced death receptor endocytosis, thus cell sensitivity to TRAIL can be associated with post-endocytic events.
Collapse
Affiliation(s)
- Artem A Artykov
- Department of Bioengineering, Institute of Bioorganic Chemistry (RAS), Moscow, Russia
| | - Anne V Yagolovich
- Department of Bioengineering, Institute of Bioorganic Chemistry (RAS), Moscow, Russia.,Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Dmitry A Dolgikh
- Department of Bioengineering, Institute of Bioorganic Chemistry (RAS), Moscow, Russia.,Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Mikhail P Kirpichnikov
- Department of Bioengineering, Institute of Bioorganic Chemistry (RAS), Moscow, Russia.,Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Daria B Trushina
- Department of X-Ray and Synchrotron Research, A.V. Shubnikov Institute of Crystallography of Federal Scientific Research Centre "Crystallography and Photonics" of Russian Academy of Sciences, Moscow, Russia
| | - Marine E Gasparian
- Department of Bioengineering, Institute of Bioorganic Chemistry (RAS), Moscow, Russia
| |
Collapse
|
3
|
From Proteomic Mapping to Invasion-Metastasis-Cascade Systemic Biomarkering and Targeted Drugging of Mutant BRAF-Dependent Human Cutaneous Melanomagenesis. Cancers (Basel) 2021; 13:cancers13092024. [PMID: 33922182 PMCID: PMC8122743 DOI: 10.3390/cancers13092024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/09/2021] [Accepted: 04/20/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Despite the recent advances in human malignancy therapy, metastasis and chemoresistance remain the principal causes of cancer-derived deaths. Given the fatal forms of cutaneous metastatic melanoma, we herein employed primary (WM115) and metastatic (WM266-4) melanoma cells, both obtained from the same patient, to identify novel biomarkers and therapeutic agents. Through state-of-the-art technologies including deep proteome landscaping, immunofluorescence phenotyping, and drug toxicity screening, we were able to describe new molecular programs, oncogenic drivers, and drug regimens, controlling the invasion-metastasis cascade during BRAFV600D-dependent melanomagenesis. It proved that proteomic navigation could foster the development of systemic biomarkering and targeted drugging for successful treatment of advanced disease. Abstract Melanoma is classified among the most notoriously aggressive human cancers. Despite the recent progress, due to its propensity for metastasis and resistance to therapy, novel biomarkers and oncogenic molecular drivers need to be promptly identified for metastatic melanoma. Hence, by employing nano liquid chromatography-tandem mass spectrometry deep proteomics technology, advanced bioinformatics algorithms, immunofluorescence, western blotting, wound healing protocols, molecular modeling programs, and MTT assays, we comparatively examined the respective proteomic contents of WM115 primary (n = 3955 proteins) and WM266-4 metastatic (n = 6681 proteins) melanoma cells. It proved that WM115 and WM266-4 cells have engaged hybrid epithelial-to-mesenchymal transition/mesenchymal-to-epithelial transition states, with TGF-β controlling their motility in vitro. They are characterized by different signatures of SOX-dependent neural crest-like stemness and distinct architectures of the cytoskeleton network. Multiple signaling pathways have already been activated from the primary melanoma stage, whereas HIF1α, the major hypoxia-inducible factor, can be exclusively observed in metastatic melanoma cells. Invasion-metastasis cascade-specific sub-routines of activated Caspase-3-triggered apoptosis and LC3B-II-dependent constitutive autophagy were also unveiled. Importantly, WM115 and WM266-4 cells exhibited diverse drug response profiles, with epirubicin holding considerable promise as a beneficial drug for metastatic melanoma clinical management. It is the proteome navigation that enables systemic biomarkering and targeted drugging to open new therapeutic windows for advanced disease.
Collapse
|
4
|
Cardoso Alves L, Corazza N, Micheau O, Krebs P. The multifaceted role of TRAIL signaling in cancer and immunity. FEBS J 2020; 288:5530-5554. [PMID: 33215853 DOI: 10.1111/febs.15637] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 11/10/2020] [Accepted: 11/17/2020] [Indexed: 12/29/2022]
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily that can lead to the induction of apoptosis in tumor or infected cells. However, activation of TRAIL signaling may also trigger nonapoptotic pathways in cancer and in nontransformed cells, that is, immune cells. Here, we review the current knowledge on noncanonical TRAIL signaling. The biological outcomes of TRAIL signaling in immune and malignant cells are presented and explained, with a focus on the role of TRAIL for natural killer (NK) cell function. Furthermore, we highlight the technical difficulties in dissecting the precise molecular mechanisms involved in the switch between apoptotic and nonapoptotic TRAIL signaling. Finally, we discuss the consequences thereof for a therapeutic manipulation of TRAIL in cancer and possible approaches to bypass these difficulties.
Collapse
Affiliation(s)
| | - Nadia Corazza
- Institute of Pathology, University of Bern, Switzerland
| | - Olivier Micheau
- INSERM, Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France
| | | |
Collapse
|
5
|
Artykov AA, Belov DA, Shipunova VO, Trushina DB, Deyev SM, Dolgikh DA, Kirpichnikov MP, Gasparian ME. Chemotherapeutic Agents Sensitize Resistant Cancer Cells to the DR5-Specific Variant DR5-B more Efficiently than to TRAIL by Modulating the Surface Expression of Death and Decoy Receptors. Cancers (Basel) 2020; 12:cancers12051129. [PMID: 32365976 PMCID: PMC7280987 DOI: 10.3390/cancers12051129] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 12/13/2022] Open
Abstract
TRAIL is considered a promising antitumor agent because it causes apoptosis of transformed cells without affecting normal cells. However, many types of tumors are cytokine resistant, and combination therapy with various chemotherapeutic drugs is being developed to overcome the resistance. We have demonstrated that the combination of TRAIL with doxorubicin, bortezomib, and panobinostat dramatically reduced the viability of TRAIL-resistant A549 and HT-29 cells. Chemotherapy even more efficiently sensitized cells to the DR5-specific mutant variant of TRAIL DR5-B, which does not have an affinity for decoy receptors. Bortezomib and doxorubicin greatly enhanced the surface expression of the death receptors DR5 and DR4, while panobinostat increased expression of DR5 and suppressed expression of DR4 in both cell lines. All drugs increased surface expression of the decoy receptors DcR1 and DcR2. Unlike the combined treatment, if the cells were pretreated with chemotherapy for 24 h, the cytotoxic activity of TRAIL was less pronounced, while sequential treatment of cells enhanced the effectiveness of DR5-B. The same results were obtained with agonistic anti-DR5 antibodies. Thus, the effectiveness of TRAIL was rather limited due to changes in the ratio of death and decoy receptors and DR5-specific agonists may be preferred in combination antitumor therapy regimens.
Collapse
Affiliation(s)
- Artem A. Artykov
- Department of Bioengineering, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (A.A.A.); (D.A.B.); (D.A.D.); (M.P.K.)
- Faculty of Biology, M.V. Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Dmitry A. Belov
- Department of Bioengineering, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (A.A.A.); (D.A.B.); (D.A.D.); (M.P.K.)
| | - Victoria O. Shipunova
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (V.O.S.); (S.M.D.)
| | - Daria B. Trushina
- Department of X-ray and Synchrotron Research, A.V. Shubnikov Institute of Crystallography of Federal Scientific Research Centre “Crystallography and Photonics” of Russian Academy of Sciences, 119333 Moscow, Russia;
| | - Sergey M. Deyev
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (V.O.S.); (S.M.D.)
| | - Dmitry A. Dolgikh
- Department of Bioengineering, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (A.A.A.); (D.A.B.); (D.A.D.); (M.P.K.)
- Faculty of Biology, M.V. Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Mikhail P. Kirpichnikov
- Department of Bioengineering, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (A.A.A.); (D.A.B.); (D.A.D.); (M.P.K.)
- Faculty of Biology, M.V. Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Marine E. Gasparian
- Department of Bioengineering, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (A.A.A.); (D.A.B.); (D.A.D.); (M.P.K.)
- Correspondence: ; Tel.: +7-905-515-7494
| |
Collapse
|
6
|
Wang Y, Guo S, Li D, Tang Y, Li L, Su L, Liu X. YIPF2 promotes chemotherapeutic agent-mediated apoptosis via enhancing TNFRSF10B recycling to plasma membrane in non-small cell lung cancer cells. Cell Death Dis 2020; 11:242. [PMID: 32303681 PMCID: PMC7165181 DOI: 10.1038/s41419-020-2436-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 12/19/2022]
Abstract
Non-small cell lung cancer (NSCLC) is the most common histological type of lung cancer, and the identification of the apoptotic process of NSCLC is vital for its treatment. Usually, both the expression level and the cell surface level of TNFRSF10B (TNF Receptor superfamily member 10B) will increase after treatment with some chemotherapeutic agents, which plays a critical role in the apoptosis induction. However, the exact molecular mechanism underlying TNFRSF10B regulation remains largely elusive. Here, we found that TNFRSF10B, along with a vesicular trafficking regulator protein, YIPF2, were upregulated after treatment with pemetrexed (PEM) in NSCLC cells. Besides, YIPF2 increased the surface level of TNFRF10B, while YIPF2 knockdown inhibited the upregulation of TNFRSF10B and its recycling to plasma membrane. In addition, RAB8 decreased the cell surface TNFRSF10B by promoting its removing from plasma membrane to cytoplasm. Furthermore, we found that YIPF2, RAB8 and TNFRSF10B proteins interacted physically with each other. YIPF2 could further inhibit the physical interaction between TNFRSF10B and RAB8, thereby suppressing the removing of TNFRSF10B from plasma membrane to cytoplasm mediated by RAB8 and maintaining its high level on cell surface. Finally, using bioinformatics database, the YIPF2-TNFRSF10B axis was confirmed to be associated with the malignant progression of lung cancer. Taken together, we show that YIPF2 promotes chemotherapeutic agent-mediated apoptosis via enhancing TNFRSF10B recycling to plasma membrane in NSCLC cells. These findings may be beneficial for the development of potential prognostic markers of NSCLC and may provide effective treatment strategy.
Collapse
Affiliation(s)
- Yingying Wang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Sen Guo
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Dongmei Li
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Yongkang Tang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Lei Li
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Ling Su
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China.
| | - Xiangguo Liu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China.
| |
Collapse
|
7
|
Singh D, Prasad CB, Biswas D, Tewari M, Kar AG, Ansari MA, Singh S, Narayan G. TRAIL receptors are differentially regulated and clinically significant in gallbladder cancer. Pathology 2020; 52:348-358. [PMID: 32111400 DOI: 10.1016/j.pathol.2019.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/26/2019] [Accepted: 12/01/2019] [Indexed: 12/15/2022]
Abstract
Deregulation of the receptors of TNF-related apoptosis inducing ligand (TRAIL) has been reported in various cancers. In an effort to define the role of these receptors we profiled their expression in gallbladder cancer (GBC) and explored their clinical significance. Expression of TRAIL receptors' mRNA in GBC was analysed through reverse transcriptase polymerase chain reaction (RT-PCR), and protein through western blotting, immunohistochemistry and enzyme-linked immunosorbent assay (ELISA). mRNA data show frequent higher expression of TRAIL receptors in GBC samples. Death receptors DR4 and DR5 showed significant negative correlation with tumour stage, T stage and tumour grade; DcR1 transcript showed positive correlation with tumour stage, N stage, M stage and tumour grade. Similarly, IHC showed frequent positive staining for DR4, DR5 and DcR1in GBC samples. Cytoplasmic and nuclear DR4 protein showed negative correlation with T stage and tumour grade, whereas cytoplasmic DcR1 protein showed positive correlation with tumour stage and N stage. Nuclear DcR1 showed positive correlation with N stage. ELISA results showed significantly higher expression of secretory DcR1 in GBC patients. Kaplan-Meier analysis demonstrated significantly decreased mean survival of patients with positive staining of cytoplasmic DcR1. High level of death receptors identified the patients with early gallbladder cancer, whereas high DcR1 expression served as a prognostic factor for poor outcome.
Collapse
Affiliation(s)
- Deepika Singh
- Cancer Genetics Laboratory, Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Chandra Bhushan Prasad
- Cancer Genetics Laboratory, Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Dipanjan Biswas
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Mallika Tewari
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Amrita Ghosh Kar
- Department of Pathology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Mumtaz Ahmed Ansari
- Department of General Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Sunita Singh
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi, India
| | - Gopeshwar Narayan
- Cancer Genetics Laboratory, Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India.
| |
Collapse
|
8
|
Wang S, He L, Wu J, Zhou Z, Gao Y, Chen J, Shao L, Zhang Y, Zhang W. Transcriptional Profiling of Human Peripheral Blood Mononuclear Cells Identifies Diagnostic Biomarkers That Distinguish Active and Latent Tuberculosis. Front Immunol 2019; 10:2948. [PMID: 31921195 PMCID: PMC6930242 DOI: 10.3389/fimmu.2019.02948] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 12/02/2019] [Indexed: 12/11/2022] Open
Abstract
Mycobacterium tuberculosis (M. tuberculosis) infection in humans can cause active disease or latent infection. However, the factors contributing to the maintenance of latent infection vs. disease progression are poorly understood. In this study, we used a genome-wide RNA sequencing (RNA-seq) approach to identify host factors associated with M. tuberculosis infection status and a novel gene signature that can distinguish active disease from latent infection. By RNA-seq, we characterized transcriptional differences in purified protein derivative (PPD)-stimulated peripheral blood mononuclear cells (PBMCs) among three groups: patients with active tuberculosis (ATB), individuals with latent TB infection (LTBI), and TB-uninfected controls (CON). A total of 401 differentially expressed genes enabled grouping of individuals into three clusters. A validation study by quantitative real-time PCR (qRT-PCR) confirmed the differential expression of TNFRSF10C, IFNG, PGM5, EBF3, and A2ML1 between the ATB and LTBI groups. Additional clinical validation was performed to evaluate the diagnostic performance of these five biomarkers using 130 subjects. The 3-gene signature set of TNFRSF10C, EBF3, and A2ML1 enabled correct classification of 91.5% of individuals, with a high sensitivity of 86.2% and specificity of 94.9%. Diagnostic performance of the 3-gene signature set was validated using a clinical cohort of 147 subjects with suspected ATB. The sensitivity and specificity of the 3-gene set for ATB were 82.4 and 92.4%, respectively. In conclusion, we detected distinct gene expression patterns in PBMCs stimulated by PPD depending on the status of M. tuberculosis infection. Furthermore, we identified a 3-gene signature set that could distinguish ATB from LTBI, which may facilitate rapid diagnosis and treatment for more effective disease control.
Collapse
Affiliation(s)
- Sen Wang
- Department of Infectious Diseases, Institute of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Lei He
- Department of Infectious Diseases, Institute of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Wu
- Department of Infectious Diseases, Institute of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Zumo Zhou
- Department of Infectious Diseases, People's Hospital of Zhuji, Zhuji, China
| | - Yan Gao
- Department of Infectious Diseases, Institute of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiazhen Chen
- Department of Infectious Diseases, Institute of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Lingyun Shao
- Department of Infectious Diseases, Institute of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Zhang
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Wenhong Zhang
- Department of Infectious Diseases, Institute of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Heterogeneous intracellular TRAIL-receptor distribution predicts poor outcome in breast cancer patients. J Mol Med (Berl) 2019; 97:1155-1167. [PMID: 31183506 DOI: 10.1007/s00109-019-01805-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 05/22/2019] [Accepted: 05/24/2019] [Indexed: 12/20/2022]
Abstract
Upon ligand binding, plasma membrane-located TNF-related apoptosis-inducing ligand (TRAIL)-receptors 1 and 2 induce apoptosis as well as cancer-promoting signaling in cancer cells. TRAIL-R3 and TRAIL-R4 are believed to negatively regulate TRAIL-mediated apoptosis. Intracellular localization of TRAIL-receptors, as observed in many tumor cells, has been associated with oncogenic features, which are distinct from membrane-associated TRAIL-R signaling. Here, analyzing a panel of 354 breast cancer specimens, we found that an unfavorable outcome correlating with cancer-promoting properties of TRAIL-R1, TRAIL-R2, and TRAIL-R4 was most significantly defined by their intracellular distribution and mutual co-expression. A nuclear or cytoplasmic heterogeneous expression pattern correlated with markedly decreased overall survival and discriminated high-risk breast cancer patients from low-risk patients with a homogeneous distribution of expression, i.e., nuclear and cytoplasmic expression. The homogeneous TRAIL-R expression was associated with favorable breast cancer surrogate markers corresponding with excellent survival prognoses at 5 years after diagnosis (hazard ratio, 0.043) and over the complete course of follow-up (hazard ratio, 0.098; both p < 0.001). No associations with specific intrinsic breast cancer subtypes were found. Our data suggest that the determination of intracellular co-expression patterns of TRAIL-R1, TRAIL-R2, and TRAIL-R4 provides an innovative and robust method for risk stratification in breast cancer patients beyond conventional prognostic markers. KEY MESSAGES: A total of 70% of breast cancer specimens show comparably high levels of intracellular TRAIL-Rs. Nuclear or cytoplasmic TRAIL-R co-expression occurs in the majority of tumors. A total of 25% of tumors show a heterogeneous expression of cytoplasmic or nuclear TRAIL-Rs. Patients with a heterogeneous TRAIL-R expression present with poor prognoses. Additive TRAIL-R-based risk stratification comprises different breast cancer subtypes.
Collapse
|
10
|
Lee MW, Kim DS, Kim HR, Park HJ, Lee JW, Sung KW, Koo HH, Yoo KH. Inhibition of N-myc expression sensitizes human neuroblastoma IMR-32 cells expressing caspase-8 to TRAIL. Cell Prolif 2019; 52:e12577. [PMID: 30724400 PMCID: PMC6536445 DOI: 10.1111/cpr.12577] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/21/2018] [Accepted: 12/28/2018] [Indexed: 12/13/2022] Open
Abstract
Objectives This study aims to explore the roles of N‐myc and caspase‐8 in TRAIL‐resistant IMR‐32 cells which exhibit MYCN oncogene amplification and lack caspase‐8 expression. Materials and methods We established N‐myc–downregulated IMR‐32 cells using shRNA lentiviral particles targeting N‐myc and examined the effect the N‐myc inhibition on TRAIL susceptibility in human neuroblastoma IMR‐32 cells expressing caspase‐8. Results Cisplatin treatment in IMR‐32 cells increased the expression of death receptor 5 (DR5; TRAIL‐R2), but not other receptors, via downregulation of NF‐κB activity. However, the cisplatin‐mediated increase in DR5 failed to induce cell death following TRAIL treatment. Furthermore, interferon (IFN)‐γ pretreatment increased caspase‐8 expression in IMR‐32 cells, but cisplatin failed to trigger TRAIL cytotoxicity. We downregulated N‐myc expression in IMR‐32 cells using N‐myc–targeting shRNA. These cells showed decreased growth rate and Bcl‐2 expression accompanied by a mild collapse in the mitochondrial membrane potential as compared with those treated with scrambled shRNA. TRAIL treatment in N‐myc–negative cells expressing caspase‐8 following IFN‐γ treatment significantly triggered apoptotic cell death. Concurrent treatment with cisplatin enhanced TRAIL‐mediated cytotoxicity, which was abrogated by an additional pretreatment with DR5:Fc chimera protein. Conclusions N‐myc and caspase‐8 expressions are involved in TRAIL susceptibility in IMR‐32 cells, and the combination of treatment with cisplatin and TRAIL may serve as a promising strategy for the development of therapeutics against neuroblastoma that is controlled by N‐myc and caspase‐8 expression.
Collapse
Affiliation(s)
- Myoung Woo Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dae Seong Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hye Ryung Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyun Jin Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hong Hoe Koo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
11
|
Gong H, Cheng W, Wang Y. Tumor necrosis factor-related apoptosis-inducing ligand inhibits the growth and aggressiveness of colon carcinoma via the exogenous apoptosis signaling pathway. Exp Ther Med 2019; 17:41-50. [PMID: 30651763 PMCID: PMC6307519 DOI: 10.3892/etm.2018.6901] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 06/15/2018] [Indexed: 12/26/2022] Open
Abstract
Colon cancer is one of the most common types of gastrointestinal tumor. Previous studies have demonstrated that tumor necrosis factor-(TNF)-related apoptosis-inducing ligand (TRAIL) reduces the aggressiveness of colon cancer tumors and promotes the apoptosis of colon carcinoma cells. In the present study, the inhibitory effects of TRAIL were investigated and the potential mechanism of TRAIL-mediated apoptosis was explored in colon cancer cells. Reverse transcription-quantitative polymerase chain reaction, western blotting, immunofluorescence, immunohistochemistry, TUNEL and flow cytometry assays were used to analyze the effects of TRAIL on the growth, migration, invasion and apoptosis of colon tumor cells. In vivo experiments were performed in mice to analyze the therapeutic effects of TRAIL. The results demonstrated that TRAIL significantly suppressed the growth of colorectal tumor cells in a dose-dependent manner (0.5–2.5 mg/ml) and also promoted colon tumor cell death. The migration and invasion of colon tumor cells were inhibited by the downregulation of fibronectin, Vimentin and E-cadherin. The apoptotic rate revealed that TRAIL (2.0 mg/ml) significantly promoted the apoptosis of colon tumor cells by regulating apoptosis-related gene expression. TRAIL administration promoted the apoptosis of colon tumor cells via the exogenous apoptosis signaling pathway due to the upregulation of caspase-3, caspase-8 and nuclear factor-κB protein expression. In vivo assays revealed that TRAIL administration significantly inhibited tumor growth and promoted apoptotic body and lymphocyte infiltration, which led to increased survival in tumor-bearing mice compared with the control group. Immunohistochemistry revealed that P53 and B-cell lymphoma-2 were downregulated in TRAIL-treated tumors. In conclusion, TRAIL treatment significantly inhibited the growth and aggressiveness of colon tumors by inducing apoptosis via the exogenous apoptosis pathway, which suggests that TRAIL may be a potential anticancer agent for colon carcinoma therapy.
Collapse
Affiliation(s)
- Hongyan Gong
- Department of General Surgery, Yantaishan Hospital, Yantai, Shandong 264000, P.R. China
| | - Weicai Cheng
- Department of General Surgery, Yantaishan Hospital, Yantai, Shandong 264000, P.R. China
| | - Yong Wang
- Department of General Surgery, Yantaishan Hospital, Yantai, Shandong 264000, P.R. China
| |
Collapse
|
12
|
Radke DI, Ling Q, Häsler R, Alp G, Ungefroren H, Trauzold A. Downregulation of TRAIL-Receptor 1 Increases TGFβ Type II Receptor Expression and TGFβ Signalling Via MicroRNA-370-3p in Pancreatic Cancer Cells. Cancers (Basel) 2018; 10:399. [PMID: 30366420 PMCID: PMC6267290 DOI: 10.3390/cancers10110399] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 10/09/2018] [Indexed: 12/28/2022] Open
Abstract
The accumulation of perturbations in signalling pathways resulting in an apoptosis-insensitive phenotype is largely responsible for the desperate prognosis of patients with pancreatic ductal adenocarcinoma (PDAC). Accumulating evidence suggests that the death receptors TRAIL-R1 and TRAIL-R2 play important roles in PDAC biology by acting as either tumour suppressors through induction of cell death or tumour promoters through induction of pro-inflammatory signalling, invasion and metastasis. TRAIL-R2 can also associate with nuclear proteins and alter the maturation of micro RNAs (miRs). By genome-wide miR profiling and quantitative PCR analyses we now demonstrate that knockdown of TRAIL-R1 in PDAC cells decreased the level of mature miR-370 and led to an increased abundance of the type II receptor for transforming growth factor β (TGFβ). Transfection of cells with an artificial miR-370-3p decreased the levels of TGFβ-RII. We further show that transient expression of the miR-370 mimic decreased TGFβ1-induced expression of SERPINE1 encoding plasminogen activator-inhibitor 1 and partially relieved TGFβ1-induced growth inhibition. Moreover, stable TRAIL-R1 knockdown in Colo357 cells increased TGFβ1-induced SERPINE1 expression and this effect was partially reversed by transient expression of the miR-370 mimic. Finally, after transient knockdown of TRAIL-R1 in Panc1 cells there was a tendency towards enhanced activation of Smad2 and JNK1/2 signalling by exogenous TGFβ1. Taken together, our study reveals that TRAIL-R1 through regulation of miR-370 can decrease the sensitivity of PDAC cells to TGFβ and therefore represents a potential tumour suppressor in late-stage PDAC.
Collapse
Affiliation(s)
- David I Radke
- Institute for Experimental Cancer Research, University of Kiel, D-24105 Kiel, Germany.
| | - Qi Ling
- Institute for Experimental Cancer Research, University of Kiel, D-24105 Kiel, Germany.
- Department of Surgery, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 31000, China.
| | - Robert Häsler
- Institute of Clinical Molecular Biology, University of Kiel, D-24105 Kiel, Germany.
| | - Gökhan Alp
- Institute for Experimental Cancer Research, University of Kiel, D-24105 Kiel, Germany.
| | - Hendrik Ungefroren
- Clinic for General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, Campus Kiel, D-24105 Kiel, Germany.
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, D-23538 Lübeck, Germany.
| | - Anna Trauzold
- Institute for Experimental Cancer Research, University of Kiel, D-24105 Kiel, Germany.
- Clinic for General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, Campus Kiel, D-24105 Kiel, Germany.
| |
Collapse
|
13
|
Turner KA, Manouchehri JM, Kalafatis M. Sensitization of recombinant human tumor necrosis factor-related apoptosis-inducing ligand-resistant malignant melanomas by quercetin. Melanoma Res 2018; 28:277-285. [PMID: 29596115 PMCID: PMC6039425 DOI: 10.1097/cmr.0000000000000447] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 02/20/2018] [Indexed: 12/30/2022]
Abstract
Malignant melanoma is the most commonly diagnosed skin cancer associated with a high rate of metastasis. Low-stage melanoma is easily treated, but metastatic malignant melanoma is an extremely treatment-resistant malignancy with low survival rates. The application of recombinant human tumor necrosis factor-related apoptosis-inducing ligand (rhTRAIL) for the treatment of metastatic malignant melanoma holds considerable promise because of its selective proapoptotic activity towards cancer cells and not nontransformed cells. Unfortunately, the clinical utilization of rhTRAIL has been terminated due to the resistance of many cancer cells to undergo apoptosis in response to rhTRAIL. However, rhTRAIL-resistance can be abrogated through the cotreatment with compounds derived from 'Mother Nature' such as quercetin that can modulate cellular components responsible for rhTRAIL-resistance. Here, we show that rhTRAIL-resistant malignant melanomas are sensitized by quercetin. Quercetin action is manifested by the upregulation of rhTRAIL-binding receptors DR4 and DR5 on the surface of cancer cells and by increased rate of the proteasome-mediated degradation of the antiapoptotic protein FLIP. Our data provide for a new efficient and nontoxic treatment of malignant melanoma.
Collapse
Affiliation(s)
- Katherine A. Turner
- Department of Chemistry, Cleveland State University
- Center for Gene Regulation in Health and Disease (GRHD)
| | - Jasmine M. Manouchehri
- Department of Chemistry, Cleveland State University
- Center for Gene Regulation in Health and Disease (GRHD)
| | - Michael Kalafatis
- Department of Chemistry, Cleveland State University
- Center for Gene Regulation in Health and Disease (GRHD)
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
14
|
Gundlach JP, Hauser C, Schlegel FM, Böger C, Röder C, Röcken C, Becker T, Egberts JH, Kalthoff H, Trauzold A. Cytoplasmic TRAIL-R1 is a positive prognostic marker in PDAC. BMC Cancer 2018; 18:777. [PMID: 30064384 PMCID: PMC6069838 DOI: 10.1186/s12885-018-4688-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 07/22/2018] [Indexed: 01/08/2023] Open
Abstract
Background The death receptors TRAIL-R1 and TRAIL-R2 are frequently overexpressed in cancer and there is an emerging evidence for their important role in malignant progression, also in the case of pancreatic ductal adenocarcinoma (PDAC). In their canonical localization at the plasma membrane, TRAIL-R1/−R2 may induce cell death and/or pro-inflammatory signaling leading to cell migration, invasion and metastasis. Although, they have repeatedly been found intracellular, in the cytoplasm and in the nucleus, their functions in intracellular locations are still not well understood. Likewise, studies dealing with the prognostic relevance of TRAIL-Rs located in particular cellular compartments are very rare. For PDAC, the correlation of nuclear TRAIL-R2 with worse patients’ prognosis has been shown recently. Corresponding data on TRAIL-R1 are not available so far. Methods In the present study we analyzed the expression of TRAIL-R1 in 106 PDACs and 28 adjacent, peritumoral non-malignant pancreatic ducts with special emphasis on its cytoplasmic and nuclear localization and correlated the immunohistochemical findings with clinico-pathological patient characteristics. Results TRAIL-R1 was found in 93.4% of all PDAC samples. Cytoplasmic staining was present with very similar intensity in tumor and normal tissue. In contrast, nuclear TRAIL-R1 staining was significantly stronger in tumor compared to normal tissue (p = 0.006). Interestingly, we found that the number of cells with cytoplasmic TRAIL-R1 staining negatively correlates with tumor grading (p = 0.043). No such correlation could be detected for nuclear TRAIL-R1. Neither, cytoplasmic nor nuclear TRAIL-R1 staining showed a correlation with other clinico-pathological parameter such as pTNM categories. However, Kaplan-Meier analyses revealed significantly prolonged median survival of patients with positive cytoplasmic TRAIL-R1 expression in more than 80% of tumor cells compared to patients with tumors containing a smaller quantity of cells positively stained for cytoplasmic TRAIL-R1 (20 vs. 8 months; p = 0.004). Conclusion Cytoplasmic TRAIL-R1 is a positive prognostic marker for patients with PDAC. Our findings indicate that loss of cytoplasmic TRAIL-R1 results in recurrent disease with more malignant phenotype thus suggesting anti-tumor activities of cytoplasmic TRAIL-R1 in PDAC.
Collapse
Affiliation(s)
- Jan-Paul Gundlach
- Department of General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, Arnold-Heller Str. 3, Haus 18, 24105, Kiel, Germany
| | - Charlotte Hauser
- Department of General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, Arnold-Heller Str. 3, Haus 18, 24105, Kiel, Germany
| | - Franka Maria Schlegel
- Institute for Experimental Cancer Research, University of Kiel, Arnold-Heller Str. 3 (Haus 17), D-24105, Kiel, Germany
| | - Christine Böger
- Department of Pathology, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, Arnold-Heller Str. 3, Haus 14, 24105, Kiel, Germany
| | - Christian Röder
- Institute for Experimental Cancer Research, University of Kiel, Arnold-Heller Str. 3 (Haus 17), D-24105, Kiel, Germany
| | - Christoph Röcken
- Department of Pathology, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, Arnold-Heller Str. 3, Haus 14, 24105, Kiel, Germany
| | - Thomas Becker
- Department of General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, Arnold-Heller Str. 3, Haus 18, 24105, Kiel, Germany
| | - Jan-Hendrik Egberts
- Department of General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, Arnold-Heller Str. 3, Haus 18, 24105, Kiel, Germany
| | - Holger Kalthoff
- Institute for Experimental Cancer Research, University of Kiel, Arnold-Heller Str. 3 (Haus 17), D-24105, Kiel, Germany
| | - Anna Trauzold
- Department of General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, Arnold-Heller Str. 3, Haus 18, 24105, Kiel, Germany. .,Institute for Experimental Cancer Research, University of Kiel, Arnold-Heller Str. 3 (Haus 17), D-24105, Kiel, Germany.
| |
Collapse
|
15
|
Ding B, Zhang W, Wu X, Wang J, Xie C, Huang X, Zhan S, Zheng Y, Huang Y, Xu N, Ding X, Gao S. DR5 mAb-conjugated, DTIC-loaded immuno-nanoparticles effectively and specifically kill malignant melanoma cells in vivo. Oncotarget 2018; 7:57160-57170. [PMID: 27494835 PMCID: PMC5302980 DOI: 10.18632/oncotarget.11014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 07/19/2016] [Indexed: 12/18/2022] Open
Abstract
We combined chemo- and immunotherapies by constructing dual therapeutic function immuno-nanoparticles (NPs) consisting of death receptor 5 monoclonal antibody (DR5 mAb)-conjugated nanoparticles loaded with dacarbazine (DTIC) (DTIC-NPs-DR5 mAb). We determined the in vivo targeting specificity of DTIC-NPs-DR5 mAb by evaluating distribution in tumor-bearing nude mice using a real-time imaging system. Therapeutic efficacy was assessed in terms of its effect on tumor volume, survival time, histomorphology, microvessel density (MVD), and apoptotic index (AI). Systemic toxicity was evaluated by measuring white blood cells (WBC) counts, alanine aminotransferase (ALT) levels, and creatinine clearance (CR).In vivo and ex vivo imaging indicates that DR5 mAb modification enhanced the accumulation of NPs within the xenograft tumor. DTIC-NPs-DR5 mAb inhibited tumor growth more effectively than DTIC or DR5 mAb alone, indicating that combining DTIC and DR5 mAb through pharmaceutical engineering achieves a better therapeutic effect. Moreover, the toxicity of DTIC-NPs-DR5 mAb was much lower than that of DTIC, implying that DR5 mAb targeting reduces nonspecific uptake of DTIC into normal tissue and thus decreases toxic side effects. These results demonstrate that DTIC-NPs-DR5 mAb is a safe and effective nanoparticle formulation with the potential to improve the efficacy and specificity of melanoma treatment.
Collapse
Affiliation(s)
- Baoyue Ding
- Department of Pharmaceutics, Medical College of Jiaxing University, Jiaxing, PR China.,Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai, PR China.,Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, USA
| | - Wei Zhang
- Department of Pharmaceutics, Shanghai Pulmonary Hospital, Tongji University, Shanghai, PR China
| | - Xin Wu
- Department of Pharmaceutics, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, PR China
| | - Jeffrey Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, USA
| | - Chen Xie
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, USA
| | - Xuan Huang
- Department of Pharmaceutics, Medical College of Jiaxing University, Jiaxing, PR China
| | - Shuyu Zhan
- Department of Pharmaceutics, Medical College of Jiaxing University, Jiaxing, PR China
| | - Yongxia Zheng
- Department of Pharmaceutics, Medical College of Jiaxing University, Jiaxing, PR China
| | - Yueyan Huang
- Department of Pharmaceutics, Medical College of Jiaxing University, Jiaxing, PR China
| | - Ningyin Xu
- Department of Pharmaceutics, Medical College of Jiaxing University, Jiaxing, PR China
| | - Xueying Ding
- Department of Pharmaceutics, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, PR China
| | - Shen Gao
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai, PR China
| |
Collapse
|
16
|
Wang JY, Liu GZ, Wilmott JS, La T, Feng YC, Yari H, Yan XG, Thorne RF, Scolyer RA, Zhang XD, Jin L. Skp2-Mediated Stabilization of MTH1 Promotes Survival of Melanoma Cells upon Oxidative Stress. Cancer Res 2017; 77:6226-6239. [PMID: 28947420 DOI: 10.1158/0008-5472.can-17-1965] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/18/2017] [Accepted: 09/18/2017] [Indexed: 11/16/2022]
Abstract
MTH1 helps prevent misincorporation of ROS-damaged dNTPs into genomic DNA; however, there is little understanding of how MTH1 itself is regulated. Here, we report that MTH1 is regulated by polyubiquitination mediated by the E3 ligase Skp2. In melanoma cells, MTH1 was upregulated commonly mainly due to its improved stability caused by K63-linked polyubiquitination. Although Skp2 along with other components of the Skp1-Cullin-F-box (SCF) ubiquitin ligase complex was physically associated with MTH1, blocking the SCF function ablated MTH1 ubiquitination and expression. Conversely, overexpressing Skp2-elevated levels of MTH1 associated with an increase in its K63-linked ubiquitination. In melanoma cell lines and patient specimens, we observed a positive correlation of Skp2 and MTH1 expression. Mechanistic investigations showed that Skp2 limited DNA damage and apoptosis triggered by oxidative stress and that MAPK upregulated Skp2 and MTH1 to render cells more resistant to such stress. Collectively, our findings identify Skp2-mediated K63-linked polyubiquitination as a critical regulatory mechanism responsible for MTH1 upregulation in melanoma, with potential implications to target the MAPK/Skp2/MTH1 pathway to improve its treatment. Cancer Res; 77(22); 6226-39. ©2017 AACR.
Collapse
Affiliation(s)
- Jia Yu Wang
- Translational Research Institute, Henan Provincial People's Hospital, Henan, China.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, New South Wales, Australia
| | - Guang Zhi Liu
- Translational Research Institute, Henan Provincial People's Hospital, Henan, China
| | - James S Wilmott
- Discipline of Pathology, The University of Sydney, and Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Ting La
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, New South Wales, Australia
| | - Yu Chen Feng
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, New South Wales, Australia
| | - Hamed Yari
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, New South Wales, Australia
| | - Xu Guang Yan
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, New South Wales, Australia
| | - Rick F Thorne
- Translational Research Institute, Henan Provincial People's Hospital, Henan, China.,School of Environmental and Life Sciences, The University of Newcastle, New South Wales, Australia
| | - Richard A Scolyer
- Discipline of Pathology, The University of Sydney, and Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Xu Dong Zhang
- Translational Research Institute, Henan Provincial People's Hospital, Henan, China. .,School of Biomedical Sciences and Pharmacy, The University of Newcastle, New South Wales, Australia
| | - Lei Jin
- Translational Research Institute, Henan Provincial People's Hospital, Henan, China. .,School of Medicine and Public Health, The University of Newcastle, New South Wales, Australia
| |
Collapse
|
17
|
Hepatitis C Virus Exploits Death Receptor 6-mediated Signaling Pathway to Facilitate Viral Propagation. Sci Rep 2017; 7:6445. [PMID: 28743875 PMCID: PMC5527075 DOI: 10.1038/s41598-017-06740-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 06/16/2017] [Indexed: 02/06/2023] Open
Abstract
The life cycle of hepatitis C virus (HCV) is highly dependent on host proteins for virus propagation. By transcriptome sequencing analysis, we identified host genes that were highly differentially expressed in HCV-infected cells. Of these candidates, we selected Death receptor 6 (DR6) for further characterization. DR6 is an orphan member of the tumor necrosis factor receptor superfamily. In the present study, we demonstrated that both mRNA and protein levels of DR6 were increased in the context of HCV replication. We further showed that promoter activity of DR6 was increased by HCV infection. By employing promoter-linked reporter assay, we showed that HCV upregulated DR6 via ROS-mediated NF-κB pathway. Both mRNA and protein levels of DR6 were increased by NS4B or NS5A. However, NS5A but not NS4B specifically interacted with DR6. We showed that HCV modulated JNK, p38 MAPK, STAT3, and Akt signaling pathways in a DR6-dependent manner. Interestingly, Akt signaling cascade was regulated by protein interplay between DR6 and NS5A. Silencing of DR6 expression resulted in decrease of infectious HCV production without affecting viral entry, replication, and translation. Together, these data indicate that HCV modulates DR6 signaling pathway for viral propagation and may contribute to HCV-mediated pathogenesis.
Collapse
|
18
|
Wang JY, Jin L, Yan XG, Sherwin S, Farrelly M, Zhang YY, Liu F, Wang CY, Guo ST, Yari H, La T, McFarlane J, Lei FX, Tabatabaee H, Chen JZ, Croft A, Jiang CC, Zhang XD. Reactive Oxygen Species Dictate the Apoptotic Response of Melanoma Cells to TH588. J Invest Dermatol 2016; 136:2277-2286. [DOI: 10.1016/j.jid.2016.06.625] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/13/2016] [Accepted: 06/20/2016] [Indexed: 12/13/2022]
|
19
|
Zhu J, Zhou Q, Tan S. Targeting miRNAs associated with surface expression of death receptors to modulate TRAIL resistance in breast cancer. Cancer Lett 2016; 383:154-160. [PMID: 27693456 DOI: 10.1016/j.canlet.2016.09.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 08/22/2016] [Accepted: 09/02/2016] [Indexed: 01/07/2023]
Abstract
Tumor necrosis factor related apoptosis-inducing ligand (TRAIL) is capable of inducing apoptosis upon engagement of its death receptors (DRs) 4 and 5. TRAIL therapy has garnered intense interest as one of the most promising agents for cancer therapy, for its selective induction of tumor-cell apoptosis while low toxicity to most normal cells. However, a variety of breast cancer cell lines could be resistant to TRAIL-induced apoptosis. Absence of DR4 and DR5 on the breast cancer cell surface has been proposed to be critically involved in resistance to TRAIL and its agonistic antibodies. Moreover, endocytosis and autophagy in breast cancer cells could induce TRAIL resistance through downregulation of surface DR4/5. MicroRNAs (miRNAs), as endogenously expressed small non-coding RNAs, function as regulators of gene expression and involve tremendous biological processes including drug resistance. In this review, we highlight recent advances in the functional role of miRNAs in endocytosis and autophagy pathways. This review aims to present that, through regulation of critical molecules involved in autophagy and endocytosis, miRNAs could lead to mislocalization of DR4/5 in breast cancer cells and therefore play an important role in TRAIL-mediated apoptosis and TRAIL resistance.
Collapse
Affiliation(s)
- Juanjuan Zhu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Qiujing Zhou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Shuhua Tan
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
20
|
Microgravity Induction of TRAIL Expression in Preosteoclast Cells Enhances Osteoclast Differentiation. Sci Rep 2016; 6:25143. [PMID: 27142480 PMCID: PMC4855152 DOI: 10.1038/srep25143] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 04/12/2016] [Indexed: 11/24/2022] Open
Abstract
Evidence indicates that astronauts experience significant bone loss in space. We previously showed that simulated microgravity (μXg) using the NASA developed rotary cell culture system (RCCS) enhanced bone resorbing osteoclast (OCL) differentiation. However, the mechanism by which μXg increases OCL formation is unclear. RANK/RANKL signaling pathway is critical for OCL differentiation. Tumor necrosis factor-related apoptosis inducing ligand (TRAIL) has been shown to increase osteoclastogenesis. We hypothesize that TRAIL may play an important role in μXg enhanced OCL differentiation. In this study, we identified by RT profiler PCR array screening that μXg induces high levels of TRAIL expression in murine preosteoclast cells in the absence of RANKL stimulation compared to ground based (Xg) cultures. We further identified that μXg elevated the adaptor protein TRAF-6 and fusion genes OC-STAMP and DC-STAMP expression in preosteoclast cells. Interestingly, neutralizing antibody against TRAIL significantly reduced μXg induced OCL formation. We further identified that over-expression of pTRAIL in RAW 264.7 cells enhanced OCL differentiation. These results indicate that TRAIL signaling plays an important role in the μXg increased OCL differentiation. Therefore, inhibition of TRAIL expression could be an effective countermeasure for μXg induced bone loss.
Collapse
|
21
|
Decreased placental and maternal serum TRAIL-R2 levels are associated with placenta accreta. Placenta 2016; 39:1-6. [DOI: 10.1016/j.placenta.2016.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 12/10/2015] [Accepted: 01/04/2016] [Indexed: 11/21/2022]
|
22
|
Ruggeri P, Cappabianca L, Farina AR, Gneo L, Mackay AR. NGF sensitizes TrkA SH-SY5Y neuroblastoma cells to TRAIL-induced apoptosis. Cell Death Discov 2016; 2:16004. [PMID: 27551499 PMCID: PMC4979468 DOI: 10.1038/cddiscovery.2016.4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 12/04/2015] [Indexed: 12/25/2022] Open
Abstract
We report a novel pro-apoptotic function for nerve growth factor (NGF) and its tropomyosin-related kinase A (TrkA) receptor in sensitizing TRAIL (TNF-related apoptotis-inducing ligand)-resistant SH-SY5Y neuroblastoma (NB) cells to TRAIL-induced apoptosis, resulting in the abrogation of anchorage-independent tumourigenic growth in vitro. We show that the TRAIL-resistant SH-SY5Y phenotype is cFLIP (cellular FLICE-like inhibitory protein) dependent and not due to low-level functional TRAIL receptor or caspase expression or an inhibitory equilibrium between functional and decoy TRAIL receptors or B-cell lymphoma 2 (Bcl-2) and BH3-only (Bcl-2 homology domain 3-only) family proteins. NGF sensitization of SH-SY5Y cells to TRAIL-induced apoptosis was dependent upon TrkA expression, activation and subsequent sequestration of cFLIP. This reduces cFLIP recruitment to TRAIL-activated death receptors and increases the recruitment of caspase-8, leading to TRAIL-induced, caspase-dependent, type II apoptosis via the intrinsic mitochondrial pathway. This effect was temporary, inhibited within 6 h by nuclear factor-κ binding (NF-κB)-mediated increase in myeloid cell leukaemia-1 (Mcl-1) expression, abrogated by transient cFLIP or B-cell lymphoma-extra large (Bcl-xL) overexpression and optimized by NF-κB and Mcl-1 inhibitors. This novel mechanism adds an important pro-apoptotic immunological dimension to NGF/TrkA interaction that may not only help to explain the association between TrkA expression, better prognosis and spontaneous remission in NB, but also provides a novel potential pro-apoptotic therapeutic use for NGF, TRAIL and inhibitors of NF-κB and/or Mcl-1 in favourable and unfavourable NBs that express TrkA and exhibit cFLIP-mediated TRAIL resistance.
Collapse
Affiliation(s)
- P Ruggeri
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, Via Vetoio, Coppito 2, L’Aquila 67100, Italy
| | - L Cappabianca
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, Via Vetoio, Coppito 2, L’Aquila 67100, Italy
| | - A R Farina
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, Via Vetoio, Coppito 2, L’Aquila 67100, Italy
| | - L Gneo
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, Via Vetoio, Coppito 2, L’Aquila 67100, Italy
| | - A R Mackay
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, Via Vetoio, Coppito 2, L’Aquila 67100, Italy
| |
Collapse
|
23
|
Duru AD, Sutlu T, Wallblom A, Uttervall K, Lund J, Stellan B, Gahrton G, Nahi H, Alici E. Deletion of Chromosomal Region 8p21 Confers Resistance to Bortezomib and Is Associated with Upregulated Decoy TRAIL Receptor Expression in Patients with Multiple Myeloma. PLoS One 2015; 10:e0138248. [PMID: 26378933 PMCID: PMC4574561 DOI: 10.1371/journal.pone.0138248] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 08/27/2015] [Indexed: 01/06/2023] Open
Abstract
Loss of the chromosomal region 8p21 negatively effects survival in patients with multiple myeloma (MM) that undergo autologous stem cell transplantation (ASCT). In this study, we aimed to identify the immunological and molecular consequences of del(8)(p21) with regards to treatment response and bortezomib resistance. In patients receiving bortezomib as a single first line agent without any high-dose therapy, we have observed that patients with del(8)(p21) responded poorly to bortezomib with 50% showing no response while patients without the deletion had a response rate of 90%. In vitro analysis revealed a higher resistance to bortezomib possibly due to an altered gene expression profile caused by del(8)(p21) including genes such as TRAIL-R4, CCDC25, RHOBTB2, PTK2B, SCARA3, MYC, BCL2 and TP53. Furthermore, while bortezomib sensitized MM cells without del(8)(p21) to TRAIL/APO2L mediated apoptosis, in cells with del(8)(p21) bortezomib failed to upregulate the pro-apoptotic death receptors TRAIL-R1 and TRAIL-R2 which are located on the 8p21 region. Also expressing higher levels of the decoy death receptor TRAIL-R4, these cells were largely resistant to TRAIL/APO2L mediated apoptosis. Corroborating the clinical outcome of the patients, our data provides a potential explanation regarding the poor response of MM patients with del(8)(p21) to bortezomib treatment. Furthermore, our clinical analysis suggests that including immunomodulatory agents such as Lenalidomide in the treatment regimen may help to overcome this negative effect, providing an alternative consideration in treatment planning of MM patients with del(8)(p21).
Collapse
Affiliation(s)
- Adil Doganay Duru
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Center for Diseases of Aging, Vaccine and Gene Therapy Institute of Florida, Port St. Lucie, Florida, United States of America
| | - Tolga Sutlu
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Nanotechnology Research and Application Center, Sabanci University, Istanbul, Turkey
| | - Ann Wallblom
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Katarina Uttervall
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Haematology Centre, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Johan Lund
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Haematology Centre, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Birgitta Stellan
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Gösta Gahrton
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Haematology Centre, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Hareth Nahi
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Haematology Centre, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Evren Alici
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Center for Diseases of Aging, Vaccine and Gene Therapy Institute of Florida, Port St. Lucie, Florida, United States of America
- Haematology Centre, Karolinska University Hospital, Huddinge, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
24
|
Expression of tumor necrosis factor-related apoptosis-inducing ligand death receptors DR4 and DR5 in human nonmelanoma skin cancer. Am J Dermatopathol 2015; 36:710-7. [PMID: 24212133 DOI: 10.1097/dad.0b013e3182a3d31d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Death receptors 4 and 5 (DR4 and DR5) are cell surface receptors that when activated by their ligand tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) triggers apoptosis in most cancer cells but not in normal cells. Currently, it remains unclear whether DR4 and DR5 are involved in immune surveillance against nonmelanoma skin cancer (NMSC) progression. The aim of this study was to investigate the expression of DR4 and DR5 in NMSC and relate the results to the established clinicopathologic prognostic factors. This study was conducted on about 80 skin specimens from patients with NMSC (40 basal cell carcinoma and 40 squamous cell carcinoma) and diagnosed and confirmed by biopsy. Immunohistochemical analysis for DR4 and DR5 was carried out on formalin-fixed paraffin-embedded sections of skin tissues using avidin-biotin peroxidase method. Significant expression of both DR4 and DR5 was observed in NMSC cases. There was statistically significant association between DR4 and DR5 expression in squamous cell carcinoma and each of tumor site and lymph node metastasis. There was statistically significant association between DR4 expression in basal cell carcinoma and histopathologic subtypes (high expression in nodular type) and between DR5 expression and tumor site (high expression in sun-exposed area). In conclusion, expression of TRAIL receptors that mediate extrinsic apoptotic pathway in NMSC may be suggestive of a reassessment of the suitability of TRAIL-based strategy in future NMSC therapies.
Collapse
|
25
|
Twomey JD, Kim SR, Zhao L, Bozza WP, Zhang B. Spatial dynamics of TRAIL death receptors in cancer cells. Drug Resist Updat 2015; 19:13-21. [PMID: 25840763 DOI: 10.1016/j.drup.2015.02.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 02/15/2015] [Accepted: 02/20/2015] [Indexed: 12/24/2022]
Abstract
TNF-related apoptosis inducing ligand (TRAIL) selectively induces apoptosis in cancer cells without harming most normal cells. Currently, multiple clinical trials are underway to evaluate the antitumor activity of recombinant human TRAIL (rhTRAIL) and agonistic antibodies that target death receptors (DRs) 4 or 5. It is encouraging that these products have shown a tolerated safety profile in early phase studies. However, their therapeutic potential is likely limited by the emergence of tumor drug resistance phenomena. Increasing evidence indicates that TRAIL DRs are deficient on the plasma membrane of some cancer cells despite their total protein expression. Notably, the lack of surface DR4/DR5 is sufficient to render cancers resistant to TRAIL-induced apoptosis, regardless of the status of other apoptosis signaling components. The current review highlights recent findings on the dynamic expression of TRAIL death receptors, including the regulatory roles of endocytosis, autophagy, and Ras GTPase-mediated signaling events. This information could aid in the identification of novel predictive biomarkers of tumor response as well as the development of combinational drugs to overcome or bypass tumor drug resistance to TRAIL receptor-targeted therapies.
Collapse
Affiliation(s)
- Julianne D Twomey
- Division of Biotechnology Review and Research IV, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States
| | - Su-Ryun Kim
- Division of Biotechnology Review and Research IV, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States
| | - Liqun Zhao
- Division of Biotechnology Review and Research IV, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States
| | - William P Bozza
- Division of Biotechnology Review and Research IV, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States
| | - Baolin Zhang
- Division of Biotechnology Review and Research IV, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States.
| |
Collapse
|
26
|
Bertsch U, Röder C, Kalthoff H, Trauzold A. Compartmentalization of TNF-related apoptosis-inducing ligand (TRAIL) death receptor functions: emerging role of nuclear TRAIL-R2. Cell Death Dis 2014; 5:e1390. [PMID: 25165876 PMCID: PMC4454323 DOI: 10.1038/cddis.2014.351] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 07/09/2014] [Accepted: 07/10/2014] [Indexed: 01/05/2023]
Abstract
Localized in the plasma membrane, death domain-containing TNF-related apoptosis-inducing ligand (TRAIL) receptors, TRAIL-R1 and TRAIL-R2, induce apoptosis and non-apoptotic signaling when crosslinked by the ligand TRAIL or by agonistic receptor-specific antibodies. Recently, an increasing body of evidence has accumulated that TRAIL receptors are additionally found in noncanonical intracellular locations in a wide range of cell types, preferentially cancer cells. Thus, besides their canonical locations in the plasma membrane and in intracellular membranes of the secretory pathway as well as endosomes and lysosomes, TRAIL receptors may also exist in autophagosomes, in nonmembraneous cytosolic compartment as well as in the nucleus. Such intracellular locations have been mainly regarded as hide-outs for these receptors representing a strategy for cancer cells to resist TRAIL-mediated apoptosis. Recently, a novel function of intracellular TRAIL-R2 has been revealed. When present in the nuclei of tumor cells, TRAIL-R2 inhibits the processing of the primary let-7 miRNA (pri-let-7) via interaction with accessory proteins of the Microprocessor complex. The nuclear TRAIL-R2-driven decrease in mature let-7 enhances the malignancy of cancer cells. This finding represents a new example of nuclear activity of typically plasma membrane-located cytokine and growth factor receptors. Furthermore, this extends the list of nucleic acid targets of the cell surface receptors by pri-miRNA in addition to DNA and mRNA. Here we review the diverse functions of TRAIL-R2 depending on its intracellular localization and we particularly discuss the nuclear TRAIL-R2 (nTRAIL-R2) function in the context of known nuclear activities of other normally plasma membrane-localized receptors.
Collapse
Affiliation(s)
- U Bertsch
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel D-24105, Germany
| | - C Röder
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel D-24105, Germany
| | - H Kalthoff
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel D-24105, Germany
| | - A Trauzold
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel D-24105, Germany
| |
Collapse
|
27
|
Bai FL, Yu YH, Tian H, Ren GP, Wang H, Zhou B, Han XH, Yu QZ, Li DS. Genetically engineered Newcastle disease virus expressing interleukin-2 and TNF-related apoptosis-inducing ligand for cancer therapy. Cancer Biol Ther 2014; 15:1226-38. [PMID: 24971746 DOI: 10.4161/cbt.29686] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Recombinant Newcastle disease virus (rNDV) have shown oncolytic therapeutic efficacy in preclinical studies and are currently in clinical trials. In this study, we have evaluated the possibility to enhance the cancer therapeutic potential of NDV by means of inserting both interleukin-2 (IL-2) and tumor necrosis factor-related apoptosis inducing ligand (TRAIL) delivered by rNDV. We demonstrated that rNDV expressing TRAIL (rNDV-TRAIL) or both human IL-2 and TRAIL (rNDV-IL-2-TRAIL) significantly enhanced inherent anti-neoplastic of rNDV by inducing apoptosis. And we showed that apoptosis-related genes mRNA expression was increased after treated with rNDV-TRAIL or rNDV-IL-2-TRAIL compared with rNDV and rNDV-IL-2. We also demonstrated that both rNDV-IL-2 and rNDV-IL-2-TRAIL induced proliferation of the CD4(+) and CD8(+) in treated mice and elicited expression of TNF-α and IFN-γ antitumor cytokines. These mice treated with oncolytic agents exhibited significant reduction in tumor development compared with mice treated with the parental virus. In addition, experiments in both hepatocellular carcinoma and melanoma-bearing mice demonstrated that the genetically engineered rNDV-IL-2-TRAIL exhibited prolonged animals' survival compared with rNDV, rNDV-IL-2, and rNDV-TRAIL. In conclusion, the immunotherapy and oncolytic virotherapy properties of NDV can be enhanced by the introduction of IL-2 and TRAIL genes, whose products initiated a broad cascade of immunological affects and induced tumor cells apoptosis in the microenvironment of the immune system.
Collapse
Affiliation(s)
- Fu-Liang Bai
- Biopharmaceutical Teaching and Research Department; College of Life Science; Northeast Agricultural University; Harbin, China
| | - Yin-Hang Yu
- Biopharmaceutical Teaching and Research Department; College of Life Science; Northeast Agricultural University; Harbin, China
| | - Hui Tian
- Biopharmaceutical Teaching and Research Department; College of Life Science; Northeast Agricultural University; Harbin, China
| | - Gui-Ping Ren
- Biopharmaceutical Teaching and Research Department; College of Life Science; Northeast Agricultural University; Harbin, China
| | - Hui Wang
- Biopharmaceutical Teaching and Research Department; College of Life Science; Northeast Agricultural University; Harbin, China
| | - Bing Zhou
- Biopharmaceutical Teaching and Research Department; College of Life Science; Northeast Agricultural University; Harbin, China
| | - Xiao-Hui Han
- Biopharmaceutical Teaching and Research Department; College of Life Science; Northeast Agricultural University; Harbin, China
| | - Qing-Zhong Yu
- USDA-ARS; Southeast Poultry Research Laboratory; Athens, GA USA
| | - De-Shan Li
- Biopharmaceutical Teaching and Research Department; College of Life Science; Northeast Agricultural University; Harbin, China; Biopharmaceutical Teaching and Research Department; College of Life Science; Northeast Agricultural University; Harbin, China
| |
Collapse
|
28
|
Yang Y, Luo Y, Li X, Yi Y. Differential expression analysis of Golgi apparatus proteomes in hepatocellular carcinomas and the surrounding liver tissues. Hepatol Res 2014; 44:542-50. [PMID: 23621634 DOI: 10.1111/hepr.12151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 04/18/2013] [Accepted: 04/23/2013] [Indexed: 02/08/2023]
Abstract
AIM Hepatocellular carcinoma (HCC) is the sixth most common malignancy worldwide. Liver is the largest human digestive gland with abundant Golgi apparatus involved in cell division, migration and apoptosis and others. METHODS In the present study, Golgi apparatus of HCC and the surrounding liver tissues were isolated by sucrose density gradient centrifugation and identified by electron microscopy and enzymology methods. Using 2-D gel electrophoresis and mass spectrometry, 17 differentially expressed protein of Golgi apparatus in HCC and the surrounding liver tissue were screened and identified in the Mascot database. RESULTS Of those differentially expressed proteins, six were upregulated and 11 were downregulated, some of them were related to the biological processes such as protein sorting, glycosylation, cell cycle regulation, transcription regulation and Golgi integrity. One protein, annexin A5, was verified to be upregulated in HCC by western blot. CONCLUSION The differentially expressed proteins may provide new insight into HCC biology and potential diagnostic and therapeutic biomarkers.
Collapse
Affiliation(s)
- Yaying Yang
- Department of Pathology, Molecular Medicine and Tumor Center, China
| | | | | | | |
Collapse
|
29
|
Hyland PL, Burke LS, Pfeiffer RM, Rotunno M, Sun D, Patil P, Wu X, Tucker MA, Goldstein AM, Yang XR. Constitutional promoter methylation and risk of familial melanoma. Epigenetics 2014; 9:685-92. [PMID: 24762620 PMCID: PMC4063827 DOI: 10.4161/epi.28151] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 02/03/2014] [Accepted: 02/07/2014] [Indexed: 12/13/2022] Open
Abstract
Constitutional epigenetic changes detected in blood or non-disease involving tissues have been associated with disease susceptibility. We measured promoter methylation of CDKN2A (p16 and p14ARF) and 13 melanoma-related genes using bisulfite pyrosequencing of blood DNA from 114 cases and 122 controls in 64 melanoma-prone families (26 segregating CDKN2A germline mutations). We also obtained gene expression data for these genes using microarrays from the same blood samples. We observed that CDKN2A epimutation is rare in melanoma families, and therefore is unlikely to cause major susceptibility in families without CDKN2A mutations. Although methylation levels for most gene promoters were very low (<5%), we observed a significantly reduced promoter methylation (odds ratio = 0.63, 95% confidence interval = 0.50, 0.80, P<0.001) and increased expression (fold change = 1.27, P = 0.048) for TNFRSF10C in melanoma cases. Future research in large prospective studies using both normal and melanoma tissues is required to assess the significance of TNFRSF10C methylation and expression changes in melanoma susceptibility.
Collapse
Affiliation(s)
- Paula L Hyland
- Division of Cancer Epidemiology and Genetics; National Cancer Institute; National Institutes of Health; Bethesda, MD USA
- Cancer Prevention Fellowship Program; Division of Cancer Prevention; National Cancer Institute; National Institutes of Health; Bethesda, MD USA
| | - Laura S Burke
- Division of Cancer Epidemiology and Genetics; National Cancer Institute; National Institutes of Health; Bethesda, MD USA
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics; National Cancer Institute; National Institutes of Health; Bethesda, MD USA
| | - Melissa Rotunno
- Division of Cancer Epidemiology and Genetics; National Cancer Institute; National Institutes of Health; Bethesda, MD USA
| | - David Sun
- Laboratory of Molecular Technology; Scientific Application International Corporation Frederick; National Cancer Institute at Frederick; Frederick, MD USA
| | - Prasad Patil
- Division of Cancer Epidemiology and Genetics; National Cancer Institute; National Institutes of Health; Bethesda, MD USA
| | - Xiaolin Wu
- Laboratory of Molecular Technology; Scientific Application International Corporation Frederick; National Cancer Institute at Frederick; Frederick, MD USA
| | - Margaret A Tucker
- Division of Cancer Epidemiology and Genetics; National Cancer Institute; National Institutes of Health; Bethesda, MD USA
| | - Alisa M Goldstein
- Division of Cancer Epidemiology and Genetics; National Cancer Institute; National Institutes of Health; Bethesda, MD USA
| | - Xiaohong Rose Yang
- Division of Cancer Epidemiology and Genetics; National Cancer Institute; National Institutes of Health; Bethesda, MD USA
| |
Collapse
|
30
|
Bickeböller M, Tagscherer KE, Kloor M, Jansen L, Chang-Claude J, Brenner H, Hoffmeister M, Toth C, Schirmacher P, Roth W, Bläker H. Functional characterization of the tumor-suppressor MARCKS in colorectal cancer and its association with survival. Oncogene 2014; 34:1150-9. [DOI: 10.1038/onc.2014.40] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 12/13/2013] [Accepted: 01/12/2014] [Indexed: 12/14/2022]
|
31
|
Noninvasive monitoring of pharmacodynamics and kinetics of a death receptor 5 antibody and its enhanced apoptosis induction in sequential application with doxorubicin. Neoplasia 2014; 15:863-74. [PMID: 23908588 DOI: 10.1593/neo.13932] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 06/10/2013] [Accepted: 06/10/2013] [Indexed: 11/18/2022]
Abstract
Induction of apoptosis plays a crucial role in the response of tumors to treatment. Thus, we investigated the pharmacodynamics and tumor saturation kinetics of a death receptor 5 antibody (anti-DR5) when combined with chemotherapeutics. For our investigations, we applied an imaging method that allows monitoring of apoptosis noninvasively in living mice. A stably transfected apoptosis reporter based on split luciferase technology facilitates to screen various chemotherapeutics and anti-DR5 on their ability to induce apoptosis in glioblastoma cells in vitro as well as in vivo. We found that doxorubicin (DOX) treatment in vitro led to significant apoptosis induction within 48 hours and to a 2.3-fold increased anti-DR5 binding to the cell surface. In contrast, cisplatin and 5-fluorouracil (5-FU) treatment altered anti-DR5 binding only marginally. Induction of apoptosis by treatment with anti-DR5 was dose- and time-dependent (both in vitro and in vivo). Simultaneous visualization of fluorescence-labeled anti-DR5 in tumor tissue and apoptosis revealed maximal apoptosis induction immediately after the compound had reached tumor site. Regarding combination therapy of anti-DR5 and DOX, we found that the sequential application of DOX before anti-DR5 resulted in synergistically enhanced apoptosis reporter activity. In striking contrast, anti-DR5 given before DOX did not lead to increased apoptosis induction. We suggest that DOX-induced recruitment of DR5 to the cell surface impacts the enhanced apoptotic effect that can be longitudinally monitored by apoptosis imaging. This study demonstrates that the combination of apoptosis and fluorescence imaging is an excellent method for optimizing dosing and treatment schedules in preclinical cancer models.
Collapse
|
32
|
Smith W, Tomasec P, Aicheler R, Loewendorf A, Nemčovičová I, Wang ECY, Stanton RJ, Macauley M, Norris P, Willen L, Ruckova E, Nomoto A, Schneider P, Hahn G, Zajonc DM, Ware CF, Wilkinson GWG, Benedict CA. Human cytomegalovirus glycoprotein UL141 targets the TRAIL death receptors to thwart host innate antiviral defenses. Cell Host Microbe 2013; 13:324-35. [PMID: 23498957 PMCID: PMC3601332 DOI: 10.1016/j.chom.2013.02.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 11/28/2012] [Accepted: 02/11/2013] [Indexed: 12/31/2022]
Abstract
Death receptors (DRs) of the TNFR superfamily contribute to antiviral immunity by promoting apoptosis and regulating immune homeostasis during infection, and viral inhibition of DR signaling can alter immune defenses. Here we identify the human cytomegalovirus (HCMV) UL141 glycoprotein as necessary and sufficient to restrict TRAIL DR function. Despite showing no primary sequence homology to TNF family cytokines, UL141 binds the ectodomains of both human TRAIL DRs with affinities comparable to the natural ligand TRAIL. UL141 binding promotes intracellular retention of the DRs, thus protecting virus infected cells from TRAIL and TRAIL-dependent NK cell-mediated killing. The identification of UL141 as a herpesvirus modulator of the TRAIL DRs strongly implicates this pathway as a regulator of host defense to HCMV and highlights UL141 as a pleiotropic inhibitor of NK cell effector function.
Collapse
Affiliation(s)
- Wendell Smith
- The La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Chen JJ, Shen HCJ, Rivera Rosado LA, Zhang Y, Di X, Zhang B. Mislocalization of death receptors correlates with cellular resistance to their cognate ligands in human breast cancer cells. Oncotarget 2013; 3:833-42. [PMID: 22909995 PMCID: PMC3478460 DOI: 10.18632/oncotarget.542] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Multiple clinical trials are ongoing to evaluate the potential antitumor activity of human TNF variants, Fas ligand (FasL), TNF-related apoptosis inducing ligand (TRAIL) and its agonistic antibodies. These drug products act through the death receptors (DRs) TNF receptor 1 (TNFR1), Fas/CD95, DR4 (TRAIL-R1) and/or DR5 (TRAIL-R2), respectively. Therefore, characterization of the level and localization of DR expression in cancer cells is important for DR-targeted therapy. In this study, we examined the subcellular distribution of the four DRs in a panel of 10 human breast cancer cell lines by western blots and flow cytometry and 50 human breast tumors by immunohistochemistry. Despite their total protein expressions, the DRs were found to be absent on the surface of some cell lines. Consistent with this result, all four DRs were found to be mostly expressed in the cytoplasm and/or the nucleus of primary breast tumors (n=50). We further determined the growth inhibition activity (GI50) of the death ligands, recombinant human TNFα, FasL and TRAIL, and found a correlation with the subcellular localization of the corresponding DRs. These results demonstrate an aberrant expression of the death receptors in breast cancer cells, and suggest that the lack of surface DRs appears to be predictive of tumor resistance to DR-targeted therapies.
Collapse
Affiliation(s)
- Jun-Jie Chen
- Division of Therapeutic Proteins, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, United States
| | | | | | | | | | | |
Collapse
|
34
|
Modulation of tumor necrosis factor related apoptosis-inducing ligand (TRAIL) receptors in a human osteoclast model in vitro. Apoptosis 2012; 17:121-31. [PMID: 21972115 DOI: 10.1007/s10495-011-0662-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
TRAIL (TNF-related apoptosis-inducing ligand) has been shown to induce apoptosis by binding to TRAIL-R1 and -R2 death receptors, but not to TRAIL-R3 or -R4, its decoy receptors that lack the internal death domain. Osteoclasts (Ocs) are sensitive to TRAIL-induced apoptosis, and modulation of these receptors may change Oc sensitivity to TRAIL. Using human Oc cultures, we first investigated the gene expression profile of these receptors (TNFRSF10 -A, -B, -C, -D encoding TRAIL-Rs 1-4) by real time PCR after adding osteotropic factors during the last week of Oc cultures. We observed a significant decrease in the expression of TNFRSF10-A after the addition of TGFβ, and an increase in that of TNFRSF10-A and -B post-PTH stimulation. Protein expression of TRAIL-R1 and -R3 was upregulated in the presence of MIP-1α, but down-regulated in the presence of TGFβ (R1), TRAIL (R2) or OPG (R3). The percentage of Ocs expressing the TRAIL-R1 and/or -R2 at their surface was increased by MIP-1α and TRAIL, increased (R2) or decreased (R1) by TGFβ, and the percentage expressing TRAIL-R3 was increased by MIP-1α, TRAIL and RANKL. Although significant, the magnitude of all these changes was of about 10-15%. While a direct correlation between these changes and TRAIL-induced Oc apoptosis was less clear, a protective effect was observed in Ocs that had been treated with OPG, and an additive effect in Ocs pre-treated with TRAIL or TGFβ increased TRAIL sensitivity.
Collapse
|
35
|
Farooqi AA, Bhatti S, Ismail M. TRAIL and vitamins: opting for keys to castle of cancer proteome instead of open sesame. Cancer Cell Int 2012; 12:22. [PMID: 22672528 PMCID: PMC3502079 DOI: 10.1186/1475-2867-12-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Accepted: 04/26/2012] [Indexed: 12/18/2022] Open
Abstract
Cancer is a multifaceted molecular disorder that is modulated by a combination of genetic, metabolic and signal transduction aberrations, which severely impair the normal homeostasis of cell growth and death. Accumulating findings highlight the fact that different genetic alterations, such as mutations in tumor suppressor genes, might be related to distinct and differential sensitivity to targeted therapies. It is becoming increasingly apparent that a multipronged approach that addresses genetic milieu (alterations in upstream and/or parallel pathways) eventually determines the response of individual tumors to therapy. Cancerous cells often acquire the ability to evade death by attenuating cell death pathways that normally function to eliminate damaged and harmful cells. Therefore impaired cell death nanomachinery and withdrawal of death receptors from cell surface are some of major determinants for the development of chemotherapeutic resistance encountered during treatment. It is therefore essential to emphasize underlying factors which predispose cells to refractoriness against TRAIL mediated cell death pathway and the relevant regulatory components involved. We bring to limelight the strategies to re-sensitize TRAIL resistant cells via vitamins to induce apoptosis.
Collapse
Affiliation(s)
- Ammad Ahmad Farooqi
- Lab for Translational Oncology and Personalized Medicine, Rashid Latif Medical College (RLMC), 35 km Ferozepur Road, Lahore, Pakistan.
| | | | | |
Collapse
|
36
|
Szegezdi E, van der Sloot AM, Mahalingam D, O'Leary L, Cool RH, Muñoz IG, Montoya G, Quax WJ, de Jong S, Samali A, Serrano L. Kinetics in signal transduction pathways involving promiscuous oligomerizing receptors can be determined by receptor specificity: apoptosis induction by TRAIL. Mol Cell Proteomics 2012; 11:M111.013730. [PMID: 22213832 PMCID: PMC3316727 DOI: 10.1074/mcp.m111.013730] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Here we show by computer modeling that kinetics and outcome of signal transduction in case of hetero-oligomerizing receptors of a promiscuous ligand largely depend on the relative amounts of its receptors. Promiscuous ligands can trigger the formation of nonproductive receptor complexes, which slows down the formation of active receptor complexes and thus can block signal transduction. Our model predicts that increasing the receptor specificity of the ligand without changing its binding parameters should result in faster receptor activation and enhanced signaling. We experimentally validated this hypothesis using the cytokine tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and its four membrane-bound receptors as an example. Bypassing ligand-induced receptor hetero-oligomerization by receptor-selective TRAIL variants enhanced the kinetics of receptor activation and augmented apoptosis. Our results suggest that control of signaling pathways by promiscuous ligands could result in apparent slow biological kinetics and blocking signal transmission. By modulating the relative amount of the different receptors for the ligand, signaling processes like apoptosis can be accelerated or decelerated and even inhibited. It also implies that more effective treatments using protein therapeutics could be achieved simply by altering specificity.
Collapse
Affiliation(s)
- Eva Szegezdi
- School of Natural Sciences, National University of Ireland, Galway, Ireland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kojima Y, Nakayama M, Nishina T, Nakano H, Koyanagi M, Takeda K, Okumura K, Yagita H. Importin β1 protein-mediated nuclear localization of death receptor 5 (DR5) limits DR5/tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL)-induced cell death of human tumor cells. J Biol Chem 2011; 286:43383-93. [PMID: 22020938 DOI: 10.1074/jbc.m111.309377] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)/death receptor 5 (DR5)-mediated cell death plays an important role in the elimination of tumor cells and transformed cells. Recently, recombinant TRAIL and agonistic anti-DR5 monoclonal antibodies have been developed and applied to cancer therapy. However, depending on the type of cancer, the sensitivity to TRAIL has been reportedly different, and some tumor cells are resistant to TRAIL-mediated apoptosis. Using confocal microscopy, we found that large amounts of DR5 were localized in the nucleus in HeLa and HepG2 cells. Moreover, these tumor cells were resistant to TRAIL, whereas DU145 cells, which do not have nuclear DR5, were highly sensitive to TRAIL. By means of immunoprecipitation and Western blot analysis, we found that DR5 and importin β1 were physically associated, suggesting that the nuclear DR5 was transported through the nuclear import pathway mediated by importin β1. Two functional nuclear localization signals were identified in DR5, the mutation of which abrogated the nuclear localization of DR5 in HeLa cells. Moreover, the nuclear transport of DR5 was also prevented by the knockdown of importin β1 using siRNA, resulting in the up-regulation of DR5 expression on the cell surface and an increased sensitivity of HeLa and HepG2 cells to TRAIL. Taken together, our findings suggest that the importin β1-mediated nuclear localization of DR5 limits the DR5/TRAIL-induced cell death of human tumor cells and thus can be a novel target to improve cancer therapy with recombinant TRAIL and anti-DR5 antibodies.
Collapse
Affiliation(s)
- Yuko Kojima
- Laboratory of Biomedical Imaging Research, Biomedical Research Center, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Ding B, Wu X, Fan W, Wu Z, Gao J, Zhang W, Ma L, Xiang W, Zhu Q, Liu J, Ding X, Gao S. Anti-DR5 monoclonal antibody-mediated DTIC-loaded nanoparticles combining chemotherapy and immunotherapy for malignant melanoma: target formulation development and in vitro anticancer activity. Int J Nanomedicine 2011; 6:1991-2005. [PMID: 21976975 PMCID: PMC3181059 DOI: 10.2147/ijn.s24094] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background The increased incidence of malignant melanoma in recent decades, along with its high mortality rate and pronounced resistance to therapy pose an enormous challenge. Novel therapeutic strategies, such as immunotherapy and targeted therapy, are urgently needed for melanoma. In this study, a new active targeting drug delivery system was constructed to combine chemotherapy and active specific immunotherapy. Methods The chemotherapeutic drug, dacarbazine (DTIC), that induces apoptosis through the intrinsic pathway which typically responds to severe DNA damage, was used as a model drug to prepare DTIC-loaded polylactic acid (PLA) nanoparticles (DTIC-NPs), which were covalently conjugated to a highly specific targeting functional TRAIL-receptor 2 (DR5) monoclonal antibody (mAb) that can contribute directly to cancer cell apoptosis or growth inhibition through the extrinsic pathway. Results Our in vitro experiments demonstrated that DTIC-PLA-DR5 mAb nanoparticles (DTIC-NPs-DR5 mAb) are an active targeting drug delivery system which can specifically target DR5-overexpressing malignant melanoma cells and become efficiently internalized. Most strikingly, compared with conventional DTIC-NPs, DTIC-NPs-DR5 mAb showed significantly enhanced cytotoxicity and increased cell apoptosis in DR5-positive malignant melanoma cells. Conclusion The DTIC-NPs-DR5 mAb described in this paper might be a potential formulation for targeting chemotherapy and immunotherapy to DR5-overexpressing metastatic melanoma.
Collapse
Affiliation(s)
- Baoyue Ding
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
TRAIL and doxorubicin combination enhances anti-glioblastoma effect based on passive tumor targeting of liposomes. J Control Release 2011; 154:93-102. [DOI: 10.1016/j.jconrel.2011.05.008] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 03/23/2011] [Accepted: 05/06/2011] [Indexed: 11/22/2022]
|
40
|
Osteoprotegerin (OPG) and Related Proteins (RANK, RANKL and TRAIL) in Thyroid Disease. World J Surg 2011; 35:1984-92. [DOI: 10.1007/s00268-011-1185-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
41
|
Increased expression of TRAIL and its death receptors DR4 and DR5 in plaque psoriasis. Arch Dermatol Res 2011; 303:389-97. [DOI: 10.1007/s00403-011-1125-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 01/09/2011] [Accepted: 01/14/2011] [Indexed: 12/18/2022]
|
42
|
Lee ALZ, Dhillon SHK, Wang Y, Pervaiz S, Fan W, Yang YY. Synergistic anti-cancer effects via co-delivery of TNF-related apoptosis-inducing ligand (TRAIL/Apo2L) and doxorubicin using micellar nanoparticles. MOLECULAR BIOSYSTEMS 2011; 7:1512-22. [DOI: 10.1039/c0mb00266f] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
43
|
TNF superfamily: a growing saga of kidney injury modulators. Mediators Inflamm 2010; 2010. [PMID: 20953353 PMCID: PMC2952810 DOI: 10.1155/2010/182958] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Revised: 08/31/2010] [Accepted: 09/06/2010] [Indexed: 12/30/2022] Open
Abstract
Members of the TNF superfamily participate in kidney disease. Tumor necrosis factor (TNF) and Fas ligand regulate renal cell survival and inflammation, and therapeutic targeting improves the outcome of experimental renal injury. TNF-related apoptosis-inducing ligand (TRAIL and its potential decoy receptor osteoprotegerin are the two most upregulated death-related genes in human diabetic nephropathy. TRAIL activates NF-kappaB in tubular cells and promotes apoptosis in tubular cells and podocytes, especially in a high-glucose environment. By contrast, osteoprotegerin plays a protective role against TRAIL-induced apoptosis. Another family member, TNF-like weak inducer of apoptosis (TWEAK induces inflammation and tubular cell death or proliferation, depending on the microenvironment. While TNF only activates canonical NF-kappaB signaling, TWEAK promotes both canonical and noncanonical NF-kappaB activation in tubular cells, regulating different inflammatory responses. TWEAK promotes the secretion of MCP-1 and RANTES through NF-kappaB RelA-containing complexes and upregulates CCl21 and CCL19 expression through NF-kappaB inducing kinase (NIK-) dependent RelB/NF-kappaB2 complexes. In vivo TWEAK promotes postnephrectomy compensatory renal cell proliferation in a noninflammatory milieu. However, in the inflammatory milieu of acute kidney injury, TWEAK promotes tubular cell death and inflammation. Therapeutic targeting of TNF superfamily cytokines, including multipronged approaches targeting several cytokines should be further explored.
Collapse
|
44
|
de Vries-van Leeuwen IJ, Kortekaas-Thijssen C, Nzigou Mandouckou JA, Kas S, Evidente A, de Boer AH. Fusicoccin-A selectively induces apoptosis in tumor cells after interferon-α priming. Cancer Lett 2010; 293:198-206. [DOI: 10.1016/j.canlet.2010.01.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Revised: 01/07/2010] [Accepted: 01/11/2010] [Indexed: 11/17/2022]
|
45
|
Cho YS, Challa S, Clancy L, Chan FKM. Lipopolysaccharide-induced expression of TRAIL promotes dendritic cell differentiation. Immunology 2010; 130:504-15. [PMID: 20406302 DOI: 10.1111/j.1365-2567.2010.03266.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Tumour necrosis factor-related apoptosis inducing ligand (TRAIL) is a death-inducing cytokine whose physiological function is not well understood. Here, we show that TRAIL has a role in programming human dendritic cell (DC) differentiation. TRAIL expression was strongly induced in DCs upon stimulation with lipopolysaccharide (LPS) or Polyinosine-polycytidylic acid (poly(I:C)) stimulation. Blockade of TRAIL with neutralizing antibody partially inhibited LPS-induced up-regulation of co-stimulatory molecules and the expression of inflammatory cytokines including interleukin-12 (IL-12) p70. In addition, neutralization of TRAIL in LPS-treated DCs inhibited the DC-driven differentiation of T cells into interferon-gamma (IFN-gamma) -producing effectors. The effects of TRAIL neutralization in poly(I:C)-treated DCs were similar, except that IL-12 production and the differentiation of effector T cells into IFN-gamma producers were not inhibited. Strikingly, TRAIL stimulation alone was sufficient to induce morphological changes resembling DC maturation, up-regulation of co-stimulatory molecules, and enhancement of DC-driven allogeneic T-cell proliferation. However, TRAIL alone did not induce inflammatory cytokine production. We further show that the effects of TRAIL on DC maturation were not the result of the induction of apoptosis, but may involve p38 activation. Hence, our data demonstrate that TRAIL co-operates with other cytokines to facilitate DC functional maturation in response to Toll-like receptor activation.
Collapse
Affiliation(s)
- Young S Cho
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | | | | | |
Collapse
|
46
|
c-Cbl-mediated degradation of TRAIL receptors is responsible for the development of the early phase of TRAIL resistance. Cell Signal 2010; 22:553-63. [PMID: 19932172 DOI: 10.1016/j.cellsig.2009.11.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2009] [Accepted: 11/13/2009] [Indexed: 12/22/2022]
Abstract
We previously reported two modes of development of acquired TRAIL resistance: early phase and late phase [1]. In these studies, we observed that greater Akt activity and the expression of Bcl-xL were related mainly to the late phase of acquired TRAIL resistance. Recently we became aware of a possible mechanism of early phase TRAIL resistance development through internalization and degradation of TRAIL receptors (DR4 and DR5). Our current studies demonstrate that TRAIL receptors rapidly diminish at the membrane as well as the cytoplasm within 4h after TRAIL exposure, but recover completely after one or two days. Our studies also reveal that Cbl, a ubiquitously expressed cytoplasmic adaptor protein, is responsible for the rapid degradation of TRAIL receptors; Cbl binds to them and induces monoubiquitination of these receptors concurrent with their degeneration soon after TRAIL exposure, creating the early phase of acquired TRAIL resistance.
Collapse
|
47
|
Oikonomou E, Kosmidou V, Katseli A, Kothonidis K, Mourtzoukou D, Kontogeorgos G, Andera L, Zografos G, Pintzas A. TRAIL receptor upregulation and the implication of KRAS/BRAF mutations in human colon cancer tumors. Int J Cancer 2009; 125:2127-2135. [PMID: 19637313 DOI: 10.1002/ijc.24613] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
TRAIL raises hopes as a promising anti-tumor agent due to its selectivity toward cancer cells. Higher expression of its pro-death receptors TRAIL-R1 (DR4) and TRAIL-R2 (DR5) attenuates higher sensitivity to TRAIL-induced apoptosis, and represents a marker for better cancer prognosis and treatment. Since receptor availability can be analogous to ligand efficacy, we performed RT-PCR analysis of DR4 and DR5 in 51 colon cancer biopsy specimens and respective normal mucosa, while 11 of these tumors were determined immunohistochemically for protein expression. Transcriptional analysis showed that DR4 and DR5 were significantly upregulated in 37 and 47% of the tumor samples respectively, while both DR4 and DR5 were coinstantaneously upregulated in 31% of the samples analyzed. Positive transcriptional regulation of DRs was recorded as early as Dukes' A stage. Furthermore, protein expression analysis yielded results comparable to DR4 and DR5 increased mRNA levels. Possible contributing events to DR upregulation involve presence of frequent oncogenic mutations in the MAPK pathway, and was investigated by direct sequencing in all 51 tumors. Samples (6/8) hosting either a KRAS(G12V) or BRAF(V600E) mutation, significantly amplified the upregulated expression of DR4 and DR5, showing strong inter-relation between overexpression and presence of oncogenic KRAS/ BRAF mutations. In the light of recent data concerning TRAIL receptor distribution, we contribute further by presenting DR5 as the most frequently upregulated DR in colon cancer. Furthermore, oncogenic mutations may directly or indirectly enhance DR expression, potentially sensitizing these tumors to TRAIL-based therapies.
Collapse
Affiliation(s)
- Eftychia Oikonomou
- Laboratory of Signal Mediated Gene Expression, Institute of Biological Research and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Nuclear and cytoplasmic death receptor 5 as prognostic factors in patients with non-small cell lung cancer treated with chemotherapy. Lung Cancer 2009; 65:98-104. [DOI: 10.1016/j.lungcan.2008.10.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2008] [Revised: 08/09/2008] [Accepted: 10/10/2008] [Indexed: 11/23/2022]
|
49
|
Ricci SB, Cerchiari U. Some relations among the dialysis membrane, metastatic cells and the immune system. Med Hypotheses 2009; 73:328-31. [PMID: 19482443 DOI: 10.1016/j.mehy.2009.03.043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2009] [Revised: 02/25/2009] [Accepted: 03/01/2009] [Indexed: 10/20/2022]
Abstract
Since results in the literature show that series of patients with kidney cancer on hemodialysis present at the postmortem examination an incidence of metastases significantly lower than such patients not on hemodialysis, the author asks what part of this favorable prognostic result is due to the dialysis membrane and what part is due to the immune system. The part due to the dialysis membrane is with every probability a blockade of metastatic cancer cells. This block, very likely consequent to phenomena of adhesion of cells to the dialysis membrane, allows a selection in that it blocks those cells that have the greatest possibility of adhesion also to the endothelium of capillaries in sites of potential colonization. The immune system would thus have, in the presence of the dialysis membrane, a greater possibility to concentrate its action on residual metastatic cells, even though uremia can have a negative influence on the clinical result. The possibility is discussed that among the various therapies of neoplasms, that for melanoma in particular may benefit from the presence of the dialysis membrane. Melanoma has demonstrated a certain sensitivity to the action of cytokines, in part also due to the presence of receptors for the latter in melanoma cells. However, the early and intense metastatic diffusion worsens the prognosis in the course of the neoplasm, and an eventual blockade, although partial, of metastatic cells could thus lead to a positive clinical result. An increase in cytokines, verified in patients on hemodialysis, does not appear to have any particular affect on the course of the disease. No signs of particular activity of the cytokines most active in an antineoplastic sense or in the sense of immune tolerance have been observed.
Collapse
Affiliation(s)
- Sante Basso Ricci
- Department of Radiotherapy, Fondazione IRCCS Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy
| | | |
Collapse
|
50
|
Jin J, Liu H, Yang C, Li G, Liu X, Qian Q, Qian W. Effective gene-viral therapy of leukemia by a new fiber chimeric oncolytic adenovirus expressing TRAIL: in vitro and in vivo evaluation. Mol Cancer Ther 2009; 8:1387-97. [PMID: 19417152 DOI: 10.1158/1535-7163.mct-08-0962] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Conditionally replicating adenoviruses (CRAd) have been under extensive investigations as anticancer agents. Previously, we found that ZD55, an adenovirus serotype 5-based CRAd, infected and killed the leukemia cells expressing coxsackie adenovirus receptor (CAR). However, majority of leukemic cells lack CAR expression on their cell surface, resulting in resistance to CRAd infection. In this study, we showed that SG235, a novel fiber chimeric CRAd that has Ad35 tropism, permitted CAR-independent cell entry, and this in turn produced selective cytopathic effects in a variety of human leukemic cells in vitro and in vivo. Moreover, SG235 expressing exogenous tumor necrosis factor-related apoptosis-inducing ligand (SG235-TRAIL) effectively induced apoptosis of leukemic cells via the activation of extrinsic and intrinsic apoptotic pathway and elicited a superior antileukemia activity compared with SG235. In addition, normal hematopoietic progenitors were resistant to the inhibitory activity of SG235 and SG235-TRAIL. Our data suggest that these novel oncolytic agents may serve as useful tools for the treatment of leukemia.
Collapse
Affiliation(s)
- Jie Jin
- Key Lab of Combined Multi-Organ Transplantation, Institute of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Ministry of Public Health, Hangzhou, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|