1
|
Guerrache A, Micheau O. TNF-Related Apoptosis-Inducing Ligand: Non-Apoptotic Signalling. Cells 2024; 13:521. [PMID: 38534365 PMCID: PMC10968836 DOI: 10.3390/cells13060521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/01/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL or Apo2 or TNFSF10) belongs to the TNF superfamily. When bound to its agonistic receptors, TRAIL can induce apoptosis in tumour cells, while sparing healthy cells. Over the last three decades, this tumour selectivity has prompted many studies aiming at evaluating the anti-tumoral potential of TRAIL or its derivatives. Although most of these attempts have failed, so far, novel formulations are still being evaluated. However, emerging evidence indicates that TRAIL can also trigger a non-canonical signal transduction pathway that is likely to be detrimental for its use in oncology. Likewise, an increasing number of studies suggest that in some circumstances TRAIL can induce, via Death receptor 5 (DR5), tumour cell motility, potentially leading to and contributing to tumour metastasis. While the pro-apoptotic signal transduction machinery of TRAIL is well known from a mechanistic point of view, that of the non-canonical pathway is less understood. In this study, we the current state of knowledge of TRAIL non-canonical signalling.
Collapse
Affiliation(s)
- Abderrahmane Guerrache
- Université de Bourgogne, 21000 Dijon, France
- INSERM Research Center U1231, «Equipe DesCarTes», 21000 Dijon, France
| | - Olivier Micheau
- Université de Bourgogne, 21000 Dijon, France
- INSERM Research Center U1231, «Equipe DesCarTes», 21000 Dijon, France
- Laboratoire d’Excellence LipSTIC, 21000 Dijon, France
| |
Collapse
|
2
|
Kotipalli A, Banerjee R, Kasibhatla SM, Joshi R. Analysis of H3K4me3-ChIP-Seq and RNA-Seq data to understand the putative role of miRNAs and their target genes in breast cancer cell lines. Genomics Inform 2021; 19:e17. [PMID: 34261302 PMCID: PMC8261273 DOI: 10.5808/gi.21020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/25/2021] [Indexed: 11/26/2022] Open
Abstract
Breast cancer is one of the leading causes of cancer in women all over the world and accounts for ~25% of newly observed cancers in women. Epigenetic modifications influence differential expression of genes through non-coding RNA and play a crucial role in cancer regulation. In the present study, epigenetic regulation of gene expression by in-silico analysis of histone modifications using chromatin immunoprecipitation sequencing (ChIP-Seq) has been carried out. Histone modification data of H3K4me3 from one normal-like and four breast cancer cell lines were used to predict miRNA expression at the promoter level. Predicted miRNA promoters (based on ChIP-Seq) were used as a probe to identify gene targets. Five triple-negative breast cancer (TNBC)‒specific miRNAs (miR153-1, miR4767, miR4487, miR6720, and miR-LET7I) were identified and corresponding 13 gene targets were predicted. Eight miRNA promoter peaks were predicted to be differentially expressed in at least three breast cancer cell lines (miR4512, miR6791, miR330, miR3180-3, miR6080, miR5787, miR6733, and miR3613). A total of 44 gene targets were identified based on the 3′-untranslated regions of downregulated mRNA genes that contain putative binding targets to these eight miRNAs. These include 17 and 15 genes in luminal-A type and TNBC respectively, that have been reported to be associated with breast cancer regulation. Of the remaining 12 genes, seven (A4GALT, C2ORF74, HRCT1, ZC4H2, ZNF512, ZNF655, and ZNF608) show similar relative expression profiles in large patient samples and other breast cancer cell lines thereby giving insight into predicted role of H3K4me3 mediated gene regulation via the miRNA-mRNA axis.
Collapse
Affiliation(s)
- Aneesh Kotipalli
- HPC-Medical and Bioinformatics Applications Group, Centre for Development of Advanced Computing, Pune 411008, India
| | - Ruma Banerjee
- HPC-Medical and Bioinformatics Applications Group, Centre for Development of Advanced Computing, Pune 411008, India
| | - Sunitha Manjari Kasibhatla
- HPC-Medical and Bioinformatics Applications Group, Centre for Development of Advanced Computing, Pune 411008, India
| | - Rajendra Joshi
- HPC-Medical and Bioinformatics Applications Group, Centre for Development of Advanced Computing, Pune 411008, India
| |
Collapse
|
3
|
Zhang S, Chen Z, Shi P, Fan S, He Y, Wang Q, Li Y, Ramalingam SS, Owonikoko TK, Sun SY. Downregulation of death receptor 4 is tightly associated with positive response of EGFR mutant lung cancer to EGFR-targeted therapy and improved prognosis. Theranostics 2021; 11:3964-3980. [PMID: 33664875 PMCID: PMC7914351 DOI: 10.7150/thno.54824] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/19/2021] [Indexed: 02/03/2023] Open
Abstract
Death receptor 4 (DR4), a cell surface receptor, mediates apoptosis or induces inflammatory cytokine secretion upon binding to its ligand depending on cell contexts. Its prognostic impact in lung cancer and connection between EGFR-targeted therapy and DR4 modulation has not been reported and thus was the focus of this study. Methods: Intracellular protein alterations were measured by Western blotting. Cell surface protein was detected with antibody staining and flow cytometry. mRNA expression was monitored with qRT-PCR. Gene transactivation was analyzed with promoter reporter assay. Drug dynamic effects in vivo were evaluated using xenografts. Gene modulations were achieved with gene overexpression and knockdown. Proteins in human archived tissues were stained with immunohistochemistry. Results: EGFR inhibitors (e.g., osimertinib) decreased DR4 levels only in EGFR mutant NSCLC cells and tumors, being tightly associated with induction of apoptosis. This modulation was lost once cells became resistant to these inhibitors. Increased levels of DR4 were detected in cell lines with acquired osimertinib resistance and in NSCLC tissues relapsed from EGFR-targeted therapy. DR4 knockdown induced apoptosis and augmented apoptosis when combined with osimertinib in both sensitive and resistant cell lines, whereas enforced DR4 expression significantly attenuated osimertinib-induced apoptosis. Mechanistically, osimertinib induced MARCH8-mediated DR4 proteasomal degradation and suppressed MEK/ERK/AP-1-dependent DR4 transcription, resulting in DR4 downregulation. Moreover, we found that DR4 positive expression in human lung adenocarcinoma was significantly associated with poor patient survival. Conclusions: Collectively, we suggest that DR4 downregulation is coupled to therapeutic efficacy of EGFR-targeted therapy and predicts improved prognosis, revealing a previously undiscovered connection between EGFR-targeted therapy and DR4 modulation.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, USA
| | - Zhen Chen
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, USA
| | - Puyu Shi
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, USA
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yong He
- Department of Respiratory Disease, Daping Hospital, Chongqing, China
| | - Qiming Wang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Yixiang Li
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, USA
| | - Suresh S. Ramalingam
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, USA
| | - Taofeek K. Owonikoko
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, USA
| | - Shi-Yong Sun
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, USA
| |
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: 74] [Impact Index Per Article: 14.8] [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
|
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
|
6
|
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
|
7
|
Pare R, Soon PS, Shah A, Lee CS. Differential expression of senescence tumour markers and its implications on survival outcomes of breast cancer patients. PLoS One 2019; 14:e0214604. [PMID: 30998679 PMCID: PMC6472879 DOI: 10.1371/journal.pone.0214604] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 03/17/2019] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is a heterogeneous disease displaying different histopathological characteristics, molecular profiling and clinical behavior. This study describes the expression patterns of senescence markers P53, DEC1 and DCR2 and assesses their significance on patient survival as a single or combined marker with P16 or P14 using breast cancer progression series. One thousand and eighty (1080) patients with primary invasive ductal carcinoma, no special type, were recruited through an 11-year retrospective study period. We constructed tissue microarrays of normal, benign hyperplasia, ductal carcinoma in situ and invasive ductal carcinoma from each patient and performed immunohistochemical staining to study the protein expression. Statistical analysis includes Pearson chi-square, Kaplan-Meier log ran test and Cox proportional hazard regression were undertaken to determine the associations and predict the survival outcomes. P53, DEC1 and DCR2 expression correlated significantly with normal, benign, premalignant and malignant tissues with (p<0.05). The expression profile of these genes increases from normal to benign to premalignant and plateaued from premalignant to malignant phenotype. There is a significant association between P53 protein expression and age, grade, staging, lymphovascular invasion, estrogen receptor, progesterone receptor and HER2 whereas DCR2 protein expression significantly correlated with tumour grade, hormone receptors status and HER2 (p<0.05 respectively). P53 overexpression correlated with increased risk of relapse (p = 0.002) specifically in patients who did not receive hormone therapy (p = 0.005) or chemotherapy (p<0.0001). The combination of P53+/P16+ is significantly correlated with poor overall and disease-free survival, whereas a combination of P53+/P14+ is associated with worse outcome in disease-free survival (p<0.05 respectively). P53 overexpression appears to be a univariate predictor of poor disease-free survival. The expression profiles of DEC1 and DCR2 do not appear to correlate with patient survival outcomes. The combination of P53 with P16, rather P53 expression alone, appears to provide more useful clinical information on patient survival outcomes in breast cancer.
Collapse
Affiliation(s)
- Rahmawati Pare
- Department of Biomedical Science and Therapeutic, Faculty of Medicine & Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Sabah Malaysia
- Ingham Institute for Applied Medical Research, Liverpool, NSW Australia
- Discipline of Pathology, School of Medicine, Western Sydney University, Liverpool, NSW Australia
- * E-mail: (RP); (CSL)
| | - Patsy S. Soon
- Ingham Institute for Applied Medical Research, Liverpool, NSW Australia
- Breast Surgery Unit, Bankstown Hospital, Bankstown, NSW Australia
- South Western Sydney Clinical School, University of New South Wales, Liverpool, NSW Australia
| | - Aashit Shah
- Breast Surgery Unit, Liverpool Hospital, Liverpool, NSW Australia
| | - Cheok Soon Lee
- Ingham Institute for Applied Medical Research, Liverpool, NSW Australia
- Discipline of Pathology, School of Medicine, Western Sydney University, Liverpool, NSW Australia
- South Western Sydney Clinical School, University of New South Wales, Liverpool, NSW Australia
- Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW Australia
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW Australia
- * E-mail: (RP); (CSL)
| |
Collapse
|
8
|
Willms A, Schittek H, Rahn S, Sosna J, Mert U, Adam D, Trauzold A. Impact of p53 status on TRAIL-mediated apoptotic and non-apoptotic signaling in cancer cells. PLoS One 2019; 14:e0214847. [PMID: 30947287 PMCID: PMC6448923 DOI: 10.1371/journal.pone.0214847] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 03/22/2019] [Indexed: 12/13/2022] Open
Abstract
Due to their ability to preferentially induce cell death in tumor cells, while sparing healthy cells, TNF-related apoptosis-inducing ligand (TRAIL) and agonistic anti-TRAIL-R1 or anti-TRAIL-R2-specific antibodies are under clinical investigations for cancer-treatment. However, TRAIL-Rs may also induce signaling pathways, which result in malignant progression. TRAIL receptors are transcriptionally upregulated via wild-type p53 following radio- or chemotherapy. Nevertheless, the impact of p53 status on the expression and signaling of TRAIL-Rs is not fully understood. Therefore, we analyzed side by side apoptotic and non-apoptotic signaling induced by TRAIL or the agonistic TRAIL-R-specific antibodies Mapatumumab (anti-TRAIL-R1) and Lexatumumab (anti-TRAIL-R2) in the two isogenic colon carcinoma cell lines HCT116 p53+/+ and p53-/-. We found that HCT116 p53+/+ cells were significantly more sensitive to TRAIL-R-triggering than p53-/- cells. Similarly, A549 lung cancer cells expressing wild-type p53 were more sensitive to TRAIL-R-mediated cell death than their derivatives with knockdown of p53. Our data demonstrate that the contribution of p53 in regulating TRAIL-R-induced apoptosis does not correlate to the levels of TRAIL-Rs at the plasma membrane, but rather to p53-mediated upregulation of Bax, favouring the mitochondrial amplification loop. Consistently, stronger caspase-9 and caspase-3 activation as well as PARP-cleavage was observed following TRAIL-R-triggering in HCT116 p53+/+ compared to HCT116 p53-/- cells. Interestingly, HCT116 p53+/+ cells showed also a more potent activation of non-canonical TRAIL-R-induced signal transduction pathways like JNK, p38 and ERK1/ERK2 than p53-/- cells. Likewise, these cells induced IL-8 expression in response to TRAIL, Mapatumumab or Lexatumumab significantly stronger than p53-/- cells. We obtained similar results in A549 cells with or without p53-knockdown and in the two isogenic colon cancer cell lines RKO p53+/+ and p53-/-. In both cellular systems, we could clearly demonstrate the potentiating effects of p53 on TRAIL-R-mediated IL-8 induction. In conclusion, we found that wild-type p53 increases TRAIL-R-mediated apoptosis but simultaneously augments non-apoptotic signaling.
Collapse
Affiliation(s)
- Anna Willms
- Division of Molecular Oncology, Institute for Experimental Cancer Research, CCC-North, University of Kiel, Kiel, Germany
| | - Hella Schittek
- Division of Molecular Oncology, Institute for Experimental Cancer Research, CCC-North, University of Kiel, Kiel, Germany
| | - Sascha Rahn
- Division of Molecular Oncology, Institute for Experimental Cancer Research, CCC-North, University of Kiel, Kiel, Germany
| | - Justyna Sosna
- Institute of Immunology, University of Kiel, Kiel, Germany
| | - Ufuk Mert
- Division of Molecular Oncology, Institute for Experimental Cancer Research, CCC-North, University of Kiel, Kiel, Germany
| | - Dieter Adam
- Institute of Immunology, University of Kiel, Kiel, Germany
| | - Anna Trauzold
- Division of Molecular Oncology, Institute for Experimental Cancer Research, CCC-North, University of Kiel, Kiel, Germany
- Clinic for General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, Kiel, Germany
- * E-mail:
| |
Collapse
|
9
|
Sun X, Cui M, Wang D, Guo B, Zhang L. Tumor necrosis factor-related apoptosis inducing ligand overexpression and Taxol treatment suppresses the growth of cervical cancer cells in vitro and in vivo. Oncol Lett 2018; 15:5744-5750. [PMID: 29556305 PMCID: PMC5844141 DOI: 10.3892/ol.2018.8071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 01/17/2018] [Indexed: 12/20/2022] Open
Abstract
Tumor necrosis factor-related apoptosis inducing ligand (TRAIL) is a member of tumor necrosis factor (TNF) superfamily and functions to promote apoptosis by binding to cell surface death receptor (DR)4 and DR5. Cancer cells are more sensitive than normal cells to TRAIL-induced apoptosis, and TRAIL-based therapeutic strategies have shown promise for the treatment of cancer. The present study investigated whether enforced overexpression of TRAIL in cervical cancer cells promoted cell death in the presence or absence of Taxol, an important first-line cancer chemotherapeutic drug. Hela human cervical cancer cells were transfected with a TRAIL expression plasmid, and the effects of the combination treatment with Taxol on apoptosis was investigated in vitro and in tumor xenografts in vivo. The results indicated that Taxol treatment and TRAIL overexpression enhanced apoptosis compared with either treatment alone. The present data indicate that Taxol may enhance the pro-apoptotic effects of TRAIL overexpression in HeLa cells by increasing cleaved caspase-3 and DR5 expression levels and decreasing Bcl-2 expression levels. Furthermore, the findings suggest a possible novel treatment option for cervical cancer and uncovers a potential mechanism of the enhancing effects of Taxol on TRAIL-induced apoptosis.
Collapse
Affiliation(s)
- Xiaojie Sun
- Department of Plastic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Manhua Cui
- Department of Gynaecology and Obstetrics, The Second Hospital of Jilin University, Changchun, Jilin 130022, P.R. China
| | - Ding Wang
- Department of Pathophysiology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021 P.R. China
| | - Baofeng Guo
- Department of Plastic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Ling Zhang
- Department of Pathophysiology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021 P.R. China
| |
Collapse
|
10
|
Dubuisson A, Micheau O. Antibodies and Derivatives Targeting DR4 and DR5 for Cancer Therapy. Antibodies (Basel) 2017; 6:E16. [PMID: 31548531 PMCID: PMC6698863 DOI: 10.3390/antib6040016] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/16/2017] [Accepted: 10/19/2017] [Indexed: 02/07/2023] Open
Abstract
Developing therapeutics that induce apoptosis in cancer cells has become an increasingly attractive approach for the past 30 years. The discovery of tumor necrosis factor (TNF) superfamily members and more specifically TNF-related apoptosis-inducing ligand (TRAIL), the only cytokine of the family capable of eradicating selectively cancer cells, led to the development of numerous TRAIL derivatives targeting death receptor 4 (DR4) and death receptor 5 (DR5) for cancer therapy. With a few exceptions, preliminary attempts to use recombinant TRAIL, agonistic antibodies, or derivatives to target TRAIL agonist receptors in the clinic have been fairly disappointing. Nonetheless, a tremendous effort, worldwide, is being put into the development of novel strategic options to target TRAIL receptors. Antibodies and derivatives allow for the design of novel and efficient agonists. We summarize and discuss here the advantages and drawbacks of the soar of TRAIL therapeutics, from the first developments to the next generation of agonistic products, with a particular insight on new concepts.
Collapse
Affiliation(s)
- Agathe Dubuisson
- University Bourgogne Franche-Comté, INSERM, LNC UMR1231, F-21079 Dijon, France.
- CovalAb, Research Department, 11 Avenue Albert Einstein, 69100 Villeurbanne, Lyon, France.
- INSERM, UMR1231, Laboratoire d'Excellence LipSTIC, F-21079 Dijon, France.
| | - Olivier Micheau
- University Bourgogne Franche-Comté, INSERM, LNC UMR1231, F-21079 Dijon, France.
- CovalAb, Research Department, 11 Avenue Albert Einstein, 69100 Villeurbanne, Lyon, France.
- INSERM, UMR1231, Laboratoire d'Excellence LipSTIC, F-21079 Dijon, France.
| |
Collapse
|
11
|
von Karstedt S, Montinaro A, Walczak H. Exploring the TRAILs less travelled: TRAIL in cancer biology and therapy. Nat Rev Cancer 2017; 17:352-366. [PMID: 28536452 DOI: 10.1038/nrc.2017.28] [Citation(s) in RCA: 416] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The discovery that the tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) can induce apoptosis of cancer cells without causing toxicity in mice has led to the in-depth study of pro-apoptotic TRAIL receptor (TRAIL-R) signalling and the development of biotherapeutic drug candidates that activate TRAIL-Rs. The outcome of clinical trials with these TRAIL-R agonists has, however, been disappointing so far. Recent evidence indicates that many cancers, in addition to being TRAIL resistant, use the endogenous TRAIL-TRAIL-R system to their own advantage. However, novel insight on two fronts - how resistance of cancer cells to TRAIL-based pro-apoptotic therapies might be overcome, and how the pro-tumorigenic effects of endogenous TRAIL might be countered - gives reasonable hope that the TRAIL system can be harnessed to treat cancer. In this Review we assess the status quo of our understanding of the biology of the TRAIL-TRAIL-R system - as well as the gaps therein - and discuss the opportunities and challenges in effectively targeting this pathway.
Collapse
Affiliation(s)
- Silvia von Karstedt
- Centre for Cell Death, Cancer and Inflammation, UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Antonella Montinaro
- Centre for Cell Death, Cancer and Inflammation, UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Henning Walczak
- Centre for Cell Death, Cancer and Inflammation, UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| |
Collapse
|
12
|
Yang J, Li G, Zhang K. Pro-survival effects by NF-κB, Akt and ERK(1/2) and anti-apoptosis actions by Six1 disrupt apoptotic functions of TRAIL-Dr4/5 pathway in ovarian cancer. Biomed Pharmacother 2016; 84:1078-1087. [PMID: 27780136 DOI: 10.1016/j.biopha.2016.10.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 10/08/2016] [Accepted: 10/09/2016] [Indexed: 01/12/2023] Open
Abstract
Apoptotic signaling provoked by death receptors, DR4 and DR5, are generally considered to promote cell death and chemosensitivity in multiple cancers, but this view is being thrown into doubt with recent findings that up-regulated DR4 and DR5 in advanced stages of ovarian cancer are associated with the poor prognosis. For this conflict, two reasonable explanations have been proposed: one is that DR4 and DR5 not exclusively mediate apoptotic pathway, but also favor survival signal; another is that apoptotic signals by DR4 and DR5 are disrupted by some regulators. This study identified these two speculations in TRAIL-resistant (SKOV-3ip1 and A2780) or sensitive (OVCAR-3) ovarian cancer cells. Activation of DR4 and DR5 using their specific ligand, TRAIL, activated pro-survival factors including NF-κB, Akt and ERK(1/2) in ovarian cancer SKOV-3ip1 and A2780 cells. Pharmacological inhibition of their activities potentiated TRAIL cytotoxicity, reducing cell viability and increasing apoptosis. Six1, a homeobox transcription factor, had higher expression in SKOV-3ip1 and A2780 cells than in OVCAR-3 cells. Silencing Six1 raised levels of apoptotic factors including cleaved Bid, caspase-8 and caspase-3, and overrode the TRAIL-resistance. Co-treatment with Six1 knockdown and peptidyl O-glycosyltransferase 14 overexpression showed additive effects on apoptosis signal, leading to increased apoptosis in SKOV-3ip1 and A2780 cells. This study demonstrated that pro-survival effects by NF-κB, Akt and ERK(1/2) and anti-apoptosis actions by Six1 disrupt apoptotic functions of TRAIL-Dr4/5 pathway in ovarian cancer, which may explain why up-regulated DR4 and DR5 in ovarian cancer are associated with poor prognosis and low survival ratio of the patients.
Collapse
Affiliation(s)
- Juan Yang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central south University, No. 126, Xian Jia Hu Road, Yue Lu District, Changsha, Hunan, 410006, China
| | - Guiyuan Li
- Cancer Research Institute, Xiangya School of Medicine, The Central South University, No 87, Xiang Ya Road, Kaifu District, Changsha, 410078, China.
| | - Keqiang Zhang
- Cancer Research Institute, Xiangya School of Medicine, The Central South University, No 87, Xiang Ya Road, Kaifu District, Changsha, 410078, China
| |
Collapse
|
13
|
de Miguel D, Lemke J, Anel A, Walczak H, Martinez-Lostao L. Onto better TRAILs for cancer treatment. Cell Death Differ 2016; 23:733-47. [PMID: 26943322 PMCID: PMC4832109 DOI: 10.1038/cdd.2015.174] [Citation(s) in RCA: 241] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 12/11/2015] [Accepted: 12/17/2015] [Indexed: 01/01/2023] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), also known as Apo-2 ligand (Apo2L), is a member of the TNF cytokine superfamily. By cross-linking TRAIL-Receptor (TRAIL-R) 1 or TRAIL-R2, also known as death receptors 4 and 5 (DR4 and DR5), TRAIL has the capability to induce apoptosis in a wide variety of tumor cells while sparing vital normal cells. The discovery of this unique property among TNF superfamily members laid the foundation for testing the clinical potential of TRAIL-R-targeting therapies in the cancer clinic. To date, two of these therapeutic strategies have been tested clinically: (i) recombinant human TRAIL and (ii) antibodies directed against TRAIL-R1 or TRAIL-R2. Unfortunately, however, these TRAIL-R agonists have basically failed as most human tumors are resistant to apoptosis induction by them. It recently emerged that this is largely due to the poor agonistic activity of these agents. Consequently, novel TRAIL-R-targeting agents with increased bioactivity are currently being developed with the aim of rendering TRAIL-based therapies more active. This review summarizes these second-generation novel formulations of TRAIL and other TRAIL-R agonists, which exhibit enhanced cytotoxic capacity toward cancer cells, thereby providing the potential of being more effective when applied clinically than first-generation TRAIL-R agonists.
Collapse
Affiliation(s)
- D de Miguel
- Departamento de Bioquímica, Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón, Zaragoza, Spain
| | - J Lemke
- UCL Cancer Institute, Faculty of Medical Sciences, University College London, London, UK
| | - A Anel
- Departamento de Bioquímica, Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón, Zaragoza, Spain
| | - H Walczak
- UCL Cancer Institute, Faculty of Medical Sciences, University College London, London, UK
| | - L Martinez-Lostao
- Departamento de Bioquímica, Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón, Zaragoza, Spain
- Instituto de Nanociencia de Aragón, Zaragoza, Spain
| |
Collapse
|
14
|
Nicholl MB, Chen X, Qin C, Bai Q, Zhu Z, Davis MR, Fang Y. IL-32α has differential effects on proliferation and apoptosis of human melanoma cell lines. J Surg Oncol 2016; 113:364-9. [DOI: 10.1002/jso.24142] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 12/08/2015] [Indexed: 12/11/2022]
Affiliation(s)
- Michael B. Nicholl
- Ellis Fischel Cancer Center; University of Missouri School of Medicine; Columbia Missouri
- South Texas Veterans Health Care System; San Antonio Texas
| | - Xuhui Chen
- Ellis Fischel Cancer Center; University of Missouri School of Medicine; Columbia Missouri
- Luohu Hospital; Shenzhen China
| | - Chenglu Qin
- Ellis Fischel Cancer Center; University of Missouri School of Medicine; Columbia Missouri
- Luohu Hospital; Shenzhen China
| | - Qian Bai
- Ellis Fischel Cancer Center; University of Missouri School of Medicine; Columbia Missouri
| | - Ziwen Zhu
- Ellis Fischel Cancer Center; University of Missouri School of Medicine; Columbia Missouri
| | - Matthew R. Davis
- Ellis Fischel Cancer Center; University of Missouri School of Medicine; Columbia Missouri
| | - Yujiang Fang
- Ellis Fischel Cancer Center; University of Missouri School of Medicine; Columbia Missouri
- Des Moines University; Des Moines Iowa
| |
Collapse
|
15
|
O'Leary L, van der Sloot AM, Reis CR, Deegan S, Ryan AE, Dhami SPS, Murillo LS, Cool RH, Correa de Sampaio P, Thompson K, Murphy G, Quax WJ, Serrano L, Samali A, Szegezdi E. Decoy receptors block TRAIL sensitivity at a supracellular level: the role of stromal cells in controlling tumour TRAIL sensitivity. Oncogene 2015; 35:1261-70. [PMID: 26050621 DOI: 10.1038/onc.2015.180] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 03/02/2015] [Accepted: 03/27/2015] [Indexed: 12/22/2022]
Abstract
Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) is a death ligand cytokine known for its cytotoxic activity against malignantly transformed cells. TRAIL induces cell death through binding to death receptors DR4 and DR5. The inhibitory decoy receptors (DcR1 and DcR2) co-expressed with death receptor 4 (DR4)/DR5 on the same cell can block the transmission of the apoptotic signal. Here, we show that DcRs also regulate TRAIL sensitivity at a supracellular level and thus represent a mechanism by which the microenvironment can diminish tumour TRAIL sensitivity. Mathematical modelling and layered or spheroid stroma-extracellular matrix-tumour cultures were used to model the tumour microenvironment. By engineering TRAIL to escape binding by DcRs, we found that DcRs do not only act in a cell-autonomous or cis-regulatory manner, but also exert trans-cellular regulation originating from stromal cells and affect tumour cells, highlighting the potent inhibitory effect of DcRs in the tumour tissue and the necessity of selective targeting of the two death-inducing TRAIL receptors to maximise efficacy.
Collapse
Affiliation(s)
- L O'Leary
- Apoptosis Research Centre, National University of Ireland, Galway, Ireland
| | - A M van der Sloot
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), Barcelona, Spain.,Institute for Research in Immunology and Cancer, University of Montreal, 2950, Chemin de Polytechnique Pavillon Marcelle-Coutu, Dock 20, Montréal, Québec, Canada
| | - C R Reis
- Department of Pharmaceutical Biology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - S Deegan
- Apoptosis Research Centre, National University of Ireland, Galway, Ireland
| | - A E Ryan
- Discipline of Pharmacology and Therapeutics, School of Medicine, National University of Ireland, Galway, Ireland
| | - S P S Dhami
- Apoptosis Research Centre, National University of Ireland, Galway, Ireland
| | - L S Murillo
- National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | - R H Cool
- Department of Pharmaceutical Biology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - P Correa de Sampaio
- Department of Oncology, University of Cambridge, Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Cambridge, UK
| | - K Thompson
- Centre for Microscopy and Imaging, National University of Ireland, Galway, Ireland
| | - G Murphy
- Department of Oncology, University of Cambridge, Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Cambridge, UK
| | - W J Quax
- Department of Pharmaceutical Biology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - L Serrano
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), Barcelona, Spain
| | - A Samali
- Apoptosis Research Centre, National University of Ireland, Galway, Ireland
| | - E Szegezdi
- Apoptosis Research Centre, National University of Ireland, Galway, Ireland
| |
Collapse
|
16
|
Sarhan D, D'Arcy P, Lundqvist A. Regulation of TRAIL-receptor expression by the ubiquitin-proteasome system. Int J Mol Sci 2014; 15:18557-73. [PMID: 25318057 PMCID: PMC4227232 DOI: 10.3390/ijms151018557] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 09/30/2014] [Accepted: 10/02/2014] [Indexed: 12/21/2022] Open
Abstract
The tumor necrosis factor (TNF)-related apoptosis-inducing ligand- receptor (TRAIL-R) family has emerged as a key mediator of cell fate and survival. Ligation of TRAIL ligand to TRAIL-R1 or TRAIL-R2 initiates the extrinsic apoptotic pathway characterized by the recruitment of death domains, assembly of the death-inducing signaling complex (DISC), caspase activation and ultimately apoptosis. Conversely the decoy receptors TRAIL-R3 and TRAIL-R4, which lack the pro-apoptotic death domain, function to dampen the apoptotic response by competing for TRAIL ligand. The tissue restricted expression of the decoy receptors on normal but not cancer cells provides a therapeutic rational for the development of selective TRAIL-mediated anti-tumor therapies. Recent clinical trials using agonistic antibodies against the apoptosis-inducing TRAIL receptors or recombinant TRAIL have been promising; however the number of patients in complete remission remains stubbornly low. The mechanisms of TRAIL resistance are relatively unexplored but may in part be due to TRAIL-R down-regulation or shedding of TRAIL-R by tumor cells. Therefore a better understanding of the mechanisms underlying TRAIL resistance is required. The ubiquitin-proteasome system (UPS) has been shown to regulate TRAIL-R members suggesting that pharmacological inhibition of the UPS may be a novel strategy to augment TRAIL-based therapies and increase efficacies. We recently identified b-AP15 as an inhibitor of proteasome deubiquitinase (DUB) activity. Interestingly, exposure of tumor cell lines to b-AP15 resulted in increased TRAIL-R2 expression and enhanced sensitivity to TRAIL-mediated apoptosis and cell death in vitro and in vivo. In conclusion, targeting the UPS may represent a novel strategy to increase the cell surface expression of pro-apoptotic TRAIL-R on cancer cells and should be considered in clinical trials targeting TRAIL-receptors in cancer patients.
Collapse
Affiliation(s)
- Dhifaf Sarhan
- Karolinska Institutet, Department of Oncology-Pathology, Stockholm S-17176, Sweden.
| | - Padraig D'Arcy
- Karolinska Institutet, Department of Oncology-Pathology, Stockholm S-17176, Sweden.
| | - Andreas Lundqvist
- Karolinska Institutet, Department of Oncology-Pathology, Stockholm S-17176, Sweden.
| |
Collapse
|
17
|
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
|
18
|
Haselmann V, Kurz A, Bertsch U, Hübner S, Olempska-Müller M, Fritsch J, Häsler R, Pickl A, Fritsche H, Annewanter F, Engler C, Fleig B, Bernt A, Röder C, Schmidt H, Gelhaus C, Hauser C, Egberts JH, Heneweer C, Rohde AM, Böger C, Knippschild U, Röcken C, Adam D, Walczak H, Schütze S, Janssen O, Wulczyn FG, Wajant H, Kalthoff H, Trauzold A. Nuclear death receptor TRAIL-R2 inhibits maturation of let-7 and promotes proliferation of pancreatic and other tumor cells. Gastroenterology 2014; 146:278-90. [PMID: 24120475 DOI: 10.1053/j.gastro.2013.10.009] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 09/02/2013] [Accepted: 10/03/2013] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Tumor necrosis factor-related apoptosis inducing ligand (TRAIL-R1) (TNFRSF10A) and TRAIL-R2 (TNFRSF10B) on the plasma membrane bind ligands that activate apoptotic and other signaling pathways. Cancer cells also might have TRAIL-R2 in the cytoplasm or nucleus, although little is known about its activities in these locations. We investigated the functions of nuclear TRAIL-R2 in cancer cell lines. METHODS Proteins that interact with TRAIL-R2 initially were identified in pancreatic cancer cells by immunoprecipitation, mass spectrometry, and immunofluorescence analyses. Findings were validated in colon, renal, lung, and breast cancer cells. Functions of TRAIL-R2 were determined from small interfering RNA knockdown, real-time polymerase chain reaction, Drosha-activity, microRNA array, proliferation, differentiation, and immunoblot experiments. We assessed the effects of TRAIL-R2 overexpression or knockdown in human pancreatic ductal adenocarcinoma (PDAC) cells and their ability to form tumors in mice. We also analyzed levels of TRAIL-R2 in sections of PDACs and non-neoplastic peritumoral ducts from patients. RESULTS TRAIL-R2 was found to interact with the core microprocessor components Drosha and DGCR8 and the associated regulatory proteins p68, hnRNPA1, NF45, and NF90 in nuclei of PDAC and other tumor cells. Knockdown of TRAIL-R2 increased Drosha-mediated processing of the let-7 microRNA precursor primary let-7 (resulting in increased levels of mature let-7), reduced levels of the let-7 targets (LIN28B and HMGA2), and inhibited cell proliferation. PDAC tissues from patients had higher levels of nuclear TRAIL-R2 than non-neoplastic pancreatic tissue, which correlated with increased nuclear levels of HMGA2 and poor outcomes. Knockdown of TRAIL-R2 in PDAC cells slowed their growth as orthotopic tumors in mice. Reduced nuclear levels of TRAIL-R2 in cultured pancreatic epithelial cells promoted their differentiation. CONCLUSIONS Nuclear TRAIL-R2 inhibits maturation of the microRNA let-7 in pancreatic cancer cell lines and increases their proliferation. Pancreatic tumor samples have increased levels of nuclear TRAIL-R2, which correlate with poor outcome of patients. These findings indicate that in the nucleus, death receptors can function as tumor promoters and might be therapeutic targets.
Collapse
Affiliation(s)
- Verena Haselmann
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel, Germany
| | - Alexandra Kurz
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel, Germany
| | - Uwe Bertsch
- Institute of Immunology, University of Kiel, Kiel, Germany
| | - Sebastian Hübner
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel, Germany
| | - Monika Olempska-Müller
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel, Germany
| | - Jürgen Fritsch
- Institute of Immunology, University of Kiel, Kiel, Germany
| | - Robert Häsler
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | - Andreas Pickl
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel, Germany
| | - Hendrik Fritsche
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel, Germany
| | - Franka Annewanter
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel, Germany
| | - Christine Engler
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel, Germany
| | - Barbara Fleig
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel, Germany
| | - Alexander Bernt
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel, Germany
| | - Christian Röder
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel, Germany
| | | | | | - Charlotte Hauser
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel, Germany; Clinic for General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University of Kiel, Kiel, Germany
| | - Jan-Hendrik Egberts
- Clinic for General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University of Kiel, Kiel, Germany
| | - Carola Heneweer
- Clinic for Diagnostic Radiology, University of Kiel, Kiel, Germany
| | - Anna Maria Rohde
- Center for Anatomy, Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Uwe Knippschild
- Department of General, Visceral and Transplantation Surgery, Centre of Surgery, University of Ulm, Ulm, Germany
| | | | - Dieter Adam
- Institute of Immunology, University of Kiel, Kiel, Germany
| | - Henning Walczak
- Centre for Cell Death, Cancer and Inflammation, University College London Cancer Institute, London, United Kingdom
| | - Stefan Schütze
- Institute of Immunology, University of Kiel, Kiel, Germany
| | - Ottmar Janssen
- Institute of Immunology, University of Kiel, Kiel, Germany
| | - F Gregory Wulczyn
- Center for Anatomy, Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Holger Kalthoff
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel, Germany
| | - Anna Trauzold
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel, Germany.
| |
Collapse
|
19
|
Edgünlü TG, Ozge A, Yalın OÖ, Kul S, Erdal ME. A Study of the Impact of Death Receptor 4 (DR4) Gene Polymorphisms in Alzheimer's Disease. Balkan Med J 2013; 30:268-72. [PMID: 25207117 DOI: 10.5152/balkanmedj.2013.7455] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 04/07/2013] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Excessive apoptosis is believed to play a role in many degenerative and non-degenerative neurological diseases including Alzheimer's disease (AD). Much recent data suggest that apoptotic mechanisms may represent the missing link between Aβ deposition and proteolysis of tau protein. However, there is emerging evidence that apoptotic mechanisms may play a role in Alzheimer's Disease pathogenesis in the absence of overt apoptosis. TNF-related apoptosis inducing ligand receptor 1 (Death Receptor 4, DR4) might impair the apoptotic signal transduction and lead to dysregulation of the homeostasis between cell survival and cell death. AIMS The aim of our study was to further investigate the relationship between genetic variants of DR4 and Alzheimer's Disease. STUDY DESIGN Case control study. METHODS Sixty-eight patients with AD were included in the study. The control group comprised 72 subjects without signs of neurodegenerative diseases, as evidenced by the examination.DNA was extracted from whole blood using the salting-out procedure. Genotypes were identified by restriction fragment length polymorphism analysis of polymerase chain reaction (PCR-RFLP) products. RESULTS We observed significant differences in the genotypic distribution of the rs6557634 polymorphism in AD patients compared with controls (p<0.05); our data suggest that the GA genotype in rs6557634 could be protective against AD (p<0.05). However, there were no significant differences between AD patients and control groups in terms of the DR4 rs20575 polymorphism (p>0.05) and the DR4 rs20576 polymorphism (p>0.05). According to haplotype analysis of the DR4 gene for rs6557634, rs20575 and rs20576 polymorphisms, GCA and GCC haplotypes might be a risk factor for AD. Also, we have shown that ACA, GGC and GGA haplotypes might be protective factors against AD. CONCLUSION The present results indicate for the first time the possible contribution of the DR4 gene rs6557634, rs20575, rs20576 polymorphisms in Alzheimer's Disease, which may influence susceptibility to Alzheimer's Disease.
Collapse
Affiliation(s)
| | - Aynur Ozge
- Department of Neurology, Mersin University School of Medicine, Mersin, Turkey
| | - Osman Özgür Yalın
- Department of Neurology, Mersin University School of Medicine, Mersin, Turkey
| | - Seval Kul
- Department of Biostatistics, Gaziantep University School of Medicine, Gaziantep, Turkey
| | - Mehmet Emin Erdal
- Department of Medical Biology and Genetics, Mersin University School of Medicine, Mersin, Turkey
| |
Collapse
|
20
|
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
|
21
|
Yuan XW, Ge XQ, Sun XT, Ding YT. Intravenous administration of endothelial progenitor cells transfected with the TRAIL gene inhibits the growth of tumors derived from H22 cells in nude mice. Shijie Huaren Xiaohua Zazhi 2012; 20:2986-2991. [DOI: 10.11569/wcjd.v20.i31.2986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the influence of intravenous administration of endothelial progenitor cells (EPCs) transfected with the TRAIL gene on the growth of tumors derived from subcutaneously inoculated H22 cells in nude mice to provide a theoretical basis for the treatment of liver cancer.
METHODS: The TRAIL gene was amplified by PCR, cloned into the pcDNA3.1 vector, and transfected into EPCs. The expression of TRAIL protein was detected by Western blot. Mice were inoculated subcutaneously with H22 cells to induce tumor formation. Tumor-bearing mice were randomly divided into three groups and injected via the tail vein with EPCs transfected with the recombinant adenoviral vector carrying the TRAIL gene, the empty vector, and normal saline, respectively.
RESULTS: Restriction enzyme digestion and DNA sequencing analyses indicate that the recombinant plasmid was constructed successfully. TRAIL expression was detected in EPCs transfected with the recombinant adenoviral vector by Western blot. The rate of reduced tumor growth was 47.77% in mice administered with EPCs carrying the TRAIL gene. Tumor volume and weight in the experimental group (0.791 cm3 ± 0.119 cm3, 0.29 g ± 0.04 g) were significantly lower than those in the two control groups (all P < 0.05).
CONCLUSION: The recombinant plasmid carrying the TRAIL gene has been successfully constructed. Intravenous administration of endothelial progenitor cells transfected with the TRAIL gene inhibits the growth of tumors derived from H22 cells in nude mice.
Collapse
|
22
|
Labovsky V, Vallone VBF, Martinez LM, Otaegui J, Chasseing NA. Expression of osteoprotegerin, receptor activator of nuclear factor kappa-B ligand, tumor necrosis factor-related apoptosis-inducing ligand, stromal cell-derived factor-1 and their receptors in epithelial metastatic breast cancer cell lines. Cancer Cell Int 2012; 12:29. [PMID: 22709548 PMCID: PMC3478192 DOI: 10.1186/1475-2867-12-29] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 06/18/2012] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND While breast cancer (BC) is the major cause of death among women worldwide, there is no guarantee of better patient survival because many of these patients develop primarily metastases, despite efforts to detect it in its early stages. Bone metastasis is a common complication that occurs in 65-80 % of patients with disseminated disease, but the molecular basis underlying dormancy, dissemination and establishment of metastasis is not understood. Our objective has been to evaluate simultaneously osteoprotegerin (OPG), receptor activator of nuclear factor kappa B ligand (RANKL), tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), stromal cell-derived factor-1 (SDF-1), and their receptors (R) in 2 human BC cell lines, MDA-MB-231 and MCF-7. METHODS OPG, RANKL, TRAIL and SDF-1 expression and release, in addition to the expression of their receptors has been investigated using immunofluorescence, immunocytochemistry and ELISA analyses. RESULTS MCF-7 cells released higher levels of OPG in conditioned media (CM) than MDA-MB-231 cells; 100 % of both types of cell expressed OPG, RANKL, TRAIL and SDF-1. Moreover, 100 % in both lines expressed membrane RANKL and RANK, whereas only 50 % expressed CXCR4. Furthermore, 100 % expressed TRAIL-R1 and R4, 30-50 % TRAIL-R2, and 40-55 % TRAIL-R3. CONCLUSIONS MCF-7 and MDA-MB-231 cells not only released OPG, but expressed RANKL, TRAIL and SDF-1. The majority of the cells also expressed RANK, CXCR4 and TRAIL-R. Since these ligands and their receptors are implicated in the regulation of proliferation, survival, migration and future bone metastasis during breast tumor progression, assessment of these molecules in tumor biopsies of BC patients could be useful in identifying patients with more aggressive tumors that are also at risk of bone metastasis, which may thus improve the available options for therapeutic intervention.
Collapse
Affiliation(s)
- Vivian Labovsky
- Laboratorio de Inmuno-Hematología, Instituto de Biología y Medicina Experimental (IBYME), Buenos Aires, Argentina.
| | | | | | | | | |
Collapse
|
23
|
Yerbes R, Palacios C, López-Rivas A. The therapeutic potential of TRAIL receptor signalling in cancer cells. Clin Transl Oncol 2012; 13:839-47. [PMID: 22126726 DOI: 10.1007/s12094-011-0744-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In tumour cells, activation of the apoptotic machinery by death receptor ligands of the tumour necrosis factor (TNF) receptor superfamily of cytokines represents a novel therapeutic strategy. However, systemic treatment of tumours with TNF-α and CD95 ligand may produce severe toxic effects. The tumour necrosis-related apoptosisinducing ligand (TRAIL) is a member of the TNF family capable of inducing apoptosis in a wide variety of cancer cells upon binding to pro-apoptotic receptors, while having no effect on the majority of normal human cells tested. Interestingly, preclinical studies in mice and nonhuman primates showed no systemic cytotoxicity upon injection of either recombinant TRAIL or agonistic TRAIL-receptor antibodies. Furthermore, these treatments have been shown to effectively suppress the growth of a range of tumour xenografts. Although unwanted effects of some TRAIL preparations have been reported in normal cells, the use of TRAIL receptor agonists could represent a suitable approach in cancer therapy. Here, we shall review our current understanding of apoptotic and non-apoptotic TRAIL signalling, the therapeutic potential of TRAIL-based approaches in cancer treatment, and the results of phase 1 and 2 clinical trials with recombinant TRAIL or agonistic TRAIL receptor antibodies, either as monotherapy or in combination with other chemotherapeutic agents.
Collapse
Affiliation(s)
- R Yerbes
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Consejo Superior de Investigaciones Científicas, Sevilla, Spain
| | | | | |
Collapse
|
24
|
Fay F, McLaughlin KM, Small DM, Fennell DA, Johnston PG, Longley DB, Scott CJ. Conatumumab (AMG 655) coated nanoparticles for targeted pro-apoptotic drug delivery. Biomaterials 2011; 32:8645-53. [PMID: 21875750 DOI: 10.1016/j.biomaterials.2011.07.065] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Accepted: 07/20/2011] [Indexed: 12/14/2022]
Abstract
Colloidal nanoparticle drug delivery systems have attracted much interest for their ability to enable effective formulation and delivery of therapeutic agents. The selective delivery of these nanoparticles to the disease site can be enhanced by coating the surface of the nanoparticles with targeting moieties, such as antibodies. In this current work, we demonstrate that antibodies on the surface of the particles can also elicit key biological effects. Specifically, we demonstrate the induction of apoptosis in colorectal HCT116 cancer cells using PLGA nanoparticles coated with Conatumumab (AMG 655) death receptor 5-specific antibodies (DR5-NP). We show that DR5-NP preferentially target DR5-expressing cells and present a sufficient density of antibody paratopes to induce apoptosis via DR5, unlike free AMG 655 or non-targeted control nanoparticles. We also demonstrate that DR5-targeted nanoparticles encapsulating the cytotoxic drug camptothecin are effectively targeted to the tumour cells, thereby producing enhanced cytotoxic effects through simultaneous drug delivery and apoptosis induction. These results demonstrate that antibodies on nanoparticulate surfaces can be exploited for dual modes of action to enhance the therapeutic utility of the modality.
Collapse
Affiliation(s)
- Francois Fay
- Molecular Therapeutics, School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | | | | | | | | | | | | |
Collapse
|
25
|
Morizot A, Mérino D, Lalaoui N, Jacquemin G, Granci V, Iessi E, Lanneau D, Bouyer F, Solary E, Chauffert B, Saas P, Garrido C, Micheau O. Chemotherapy overcomes TRAIL-R4-mediated TRAIL resistance at the DISC level. Cell Death Differ 2010; 18:700-11. [PMID: 21072058 DOI: 10.1038/cdd.2010.144] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
TNF-related apoptosis-inducing ligand or Apo2L (Apo2L/TRAIL) is a promising anti-cancer drug owing to its ability to trigger apoptosis by binding to TRAIL-R1 or TRAIL-R2, two membrane-bound receptors that are often expressed by tumor cells. TRAIL can also bind non-functional receptors such as TRAIL-R4, but controversies still exist regarding their potential to inhibit TRAIL-induced apoptosis. We show here that TRAIL-R4, expressed either endogenously or ectopically, inhibits TRAIL-induced apoptosis. Interestingly, the combination of chemotherapeutic drugs with TRAIL restores tumor cell sensitivity to apoptosis in TRAIL-R4-expressing cells. This sensitization, which mainly occurs at the death-inducing signaling complex (DISC) level, through enhanced caspase-8 recruitment and activation, is compromised by c-FLIP expression and is independent of the mitochondria. Importantly, TRAIL-R4 expression prevents TRAIL-induced tumor regression in nude mice, but tumor regression induced by TRAIL can be restored with chemotherapy. Our results clearly support a negative regulatory function for TRAIL-R4 in controlling TRAIL signaling, and unveil the ability of TRAIL-R4 to cooperate with c-FLIP to inhibit TRAIL-induced cell death.
Collapse
Affiliation(s)
- A Morizot
- INSERM, U866, Dijon, F-21079 France [2] Faculty of Medicine and Pharmacy, Université de Bourgogne, Dijon, F-21079, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Yoldas B, Ozer C, Ozen O, Canpolat T, Dogan I, Griffith TS, Sanlioglu S, Ozluoglu LN. Clinical significance of TRAIL and TRAIL receptors in patients with head and neck cancer. Head Neck 2010; 33:1278-84. [PMID: 21837697 DOI: 10.1002/hed.21598] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Revised: 07/14/2010] [Accepted: 07/29/2010] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a death ligand currently under clinical trials for cancer. The molecular profile of TRAIL and TRAIL receptors has not yet been mapped for patients with laryngeal squamous cell carcinoma (SCC) or patients with oral cavity squamous cell carcinoma (OCSCC). METHODS Paraffin-embedded tissues from 60 patients with laryngeal SCC and 14 patients with OCSCC were retrospectively analyzed using immunohistochemistry. RESULTS An increase in decoy-R1 (DcR1) but a decrease in decoy-R2 (DcR2) expression were observed in patients with laryngeal SCC and in patients with OCSCC compared with control individuals with benign lesions. Clinical and pathologic grading revealed distinctive TRAIL and TRAIL receptor profiles in patients with squamous cell carcinoma of the head and neck (SCCHN). CONCLUSIONS TRAIL and a TRAIL receptor expression profile might be useful to follow-up disease progression by virtue of its connection with clinical staging and pathologic grading in patients with laryngeal SCC.
Collapse
Affiliation(s)
- Burcak Yoldas
- Human Gene Therapy Division of the Department of Medical Genetics, Akdeniz University Faculty of Medicine, Antalya, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
27
|
XVIII Annual Congress of the European Society of Gene and Cell Therapy (ESGCT)October 22–25, 2010Milan, Italy. Hum Gene Ther 2010. [DOI: 10.1089/hum.2010.915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
28
|
Loreto C, Barbagli G, Djinovic R, Vespasiani G, Carnazza ML, Miano R, Musumeci G, Sansalone S. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and its death receptor (DR5) in Peyronie's disease. A biomolecular study of apoptosis activation. J Sex Med 2010; 8:109-15. [PMID: 20840533 DOI: 10.1111/j.1743-6109.2010.02003.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Peyronie's disease (PD) is a connective tissue disorder of tunica albuginea (TA), a thick fibrous sheath surrounding the corpora cavernosa of the penis. Relatively, little is known about the disease itself. AIM To investigate whether the apoptosis cascade in degenerated and macroscopically deformed TA from men with PD is activated through the extrinsic pathway, by assessing the immunoexpression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and its death receptor, DR5. METHODS TA plaques from 15 men with PD and from four unaffected men were processed for TRAIL and DR5 immunohistochemistry and Western blot analysis. MAIN OUTCOME MEASURES A greater understanding of the pathophysiology of PD through a molecular approach, to gain insights that may lead to novel forms of treatment. RESULTS Activation of the apoptosis mechanisms through the extrinsic pathway was demonstrated by TRAIL and DR5 overexpression in fibroblasts and myofibroblasts from affected TA. CONCLUSION The finding that apoptosis activation in TA plaques occurs, at least in part, via the extrinsic pathway may help devise novel therapeutic options for these patients.
Collapse
Affiliation(s)
- Carla Loreto
- Department of Anatomy, Diagnostic Pathology, Forensic Medicine, Hygiene and Public Health, University of Catania, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Bilski A, Pasz-Walczak G, Kubiak R, Sek P, Chalubinska J, Fendler W, Wronski K, Piekarska A, Pluta P, Potemski P, Jeziorski A, Piekarski J. TRAIL protein expression in breast cancer cells correlates with nuclear grade. Arch Med Sci 2010; 6:545-51. [PMID: 22371798 PMCID: PMC3284069 DOI: 10.5114/aoms.2010.14466] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Revised: 12/15/2009] [Accepted: 01/02/2010] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION TRAIL protein may serve as an escape mechanism for cancer cells from the immune response. The aim of the study was to assess whether the presence of TRAIL protein correlates with unfavourable prognostic factors in breast carcinoma. MATERIAL AND METHODS The study group was composed of breast cancer patients treated surgically in the Department of Surgical Oncology, Medical University of Lodz, Poland, from January to December 2003. Inclusion criteria for the study were fulfilled by 117 women. The immunohistochemical study of TRAIL protein expression was performed in 118 breast carcinomas diagnosed in the study group. TRAIL protein expression was correlated with other variables: tumour size, lymph node status, grade, histological type of carcinoma, oestrogen and progesterone receptor status, HER2 expression, presence of lymphovascular invasion and age of the patient. RESULTS Expression of TRAIL protein was present in 73% of breast carcinomas. The percentage of TRAIL-expressing breast carcinoma cells correlated with the nuclear grade (τ = 0.26, p < 0.05; Tau Kendall test). The intensity of TRAIL expression (intensity of staining) in breast carcinoma cells correlated with the nuclear grade (τ = 0.15, p < 0.05; Tau Kendall test). TRAIL expression in breast carcinoma did not correlate with other studied variables. CONCLUSIONS Our analysis revealed that expression of TRAIL protein in breast carcinoma cells correlates with nuclear grade of carcinoma.
Collapse
Affiliation(s)
- Adam Bilski
- Department of Surgical Oncology, Chair of Oncology, Medical University of Lodz, Poland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Bisgin A, Terzioglu E, Aydin C, Yoldas B, Yazisiz V, Balci N, Bagci H, Gorczynski RM, Akdis CA, Sanlioglu S. TRAIL death receptor-4, decoy receptor-1 and decoy receptor-2 expression on CD8+ T cells correlate with the disease severity in patients with rheumatoid arthritis. BMC Musculoskelet Disord 2010; 11:192. [PMID: 20799941 PMCID: PMC2936350 DOI: 10.1186/1471-2474-11-192] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Accepted: 08/27/2010] [Indexed: 01/16/2023] Open
Abstract
Background Rheumatoid Arthritis (RA) is a chronic autoimmune inflammatory disorder. Although the pathogenesis of disease is unclear, it is well known that T cells play a major role in both development and perpetuation of RA through activating macrophages and B cells. Since the lack of TNF-Related Apoptosis Inducing Ligand (TRAIL) expression resulted in defective thymocyte apoptosis leading to an autoimmune disease, we explored evidence for alterations in TRAIL/TRAIL receptor expression on peripheral T lymphocytes in the molecular mechanism of RA development. Methods The expression of TRAIL/TRAIL receptors on T cells in 20 RA patients and 12 control individuals were analyzed using flow cytometry. The correlation of TRAIL and its receptor expression profile was compared with clinical RA parameters (RA activity scored as per DAS28) using Spearman Rho Analysis. Results While no change was detected in the ratio of CD4+ to CD8+ T cells between controls and RA patient groups, upregulation of TRAIL and its receptors (both death and decoy) was detected on both CD4+ and CD8+ T cells in RA patients compared to control individuals. Death Receptor-4 (DR4) and the decoy receptors DcR1 and DcR2 on CD8+ T cells, but not on CD4+ T cells, were positively correlated with patients' DAS scores. Conclusions Our data suggest that TRAIL/TRAIL receptor expression profiles on T cells might be important in revelation of RA pathogenesis.
Collapse
Affiliation(s)
- Atil Bisgin
- Department of Medical Genetics, Human Gene and Cell Therapy Center of Akdeniz University Hospitals and Clinics, Antalya, Turkey
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Dirice E, Sanlioglu AD, Kahraman S, Ozturk S, Balci MK, Omer A, Griffith TS, Sanlioglu S. Adenovirus-Mediated TRAIL Gene (Ad5hTRAIL) Delivery into Pancreatic Islets Prolongs Normoglycemia in Streptozotocin-Induced Diabetic Rats. Hum Gene Ther 2009; 20:1177-89. [DOI: 10.1089/hum.2009.039] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Ercument Dirice
- Human Gene Therapy Division, Department of Medical Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
- Department of Medical Biology and Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| | - Ahter Dilsad Sanlioglu
- Human Gene Therapy Division, Department of Medical Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
- Department of Medical Biology and Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| | - Sevim Kahraman
- Human Gene Therapy Division, Department of Medical Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
- Department of Medical Biology and Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| | - Saffet Ozturk
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| | - Mustafa Kemal Balci
- Division of Endocrinology and Metabolic Diseases, Department of Medicine, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| | - Abdulkadir Omer
- Section on Islet Transplantation and Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, U.S.A
| | | | - Salih Sanlioglu
- Human Gene Therapy Division, Department of Medical Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
- Department of Medical Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| |
Collapse
|
32
|
Abstract
Inducing apoptosis has become an important approach in the development of new anti-cancer treatments. Tumour necrosis factor apoptosis inducing ligand (TRAIL) based therapies have emerged as one of the most promising examples of this as they selectively induce apoptosis in tumour cells. However, many primary tumours are inherently resistant to TRAIL-mediated apoptosis and require additional sensitisation. Here we review apoptotic and non-apoptotic TRAIL-signalling, and the therapeutic effects of TRAIL-based treatments both as monotherapy and in combination with sensitising agents.
Collapse
Affiliation(s)
- Thomas Newsom-Davis
- Department of Immunology, Tumour Immunology Unit, Imperial College London, Hammersmith Campus, London, UK
| | | | | |
Collapse
|
33
|
Sanlioglu AD, Dirice E, Elpek O, Korcum AF, Ozdogan M, Suleymanlar I, Balci MK, Griffith TS, Sanlioglu S. High TRAIL death receptor 4 and decoy receptor 2 expression correlates with significant cell death in pancreatic ductal adenocarcinoma patients. Pancreas 2009; 38:154-160. [PMID: 18981952 DOI: 10.1097/mpa.0b013e31818db9e3] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVES The importance of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and TRAIL receptor expression in pancreatic carcinoma development is not known. To reveal the putative connection of TRAIL and TRAIL receptor expression profile to this process, we analyzed and compared the expression profile of TRAIL and its receptors in pancreatic tissues of both noncancer patients and patients with pancreatic ductal adenocarcinoma (PDAC). METHODS Thirty-one noncancer patients and 34 PDAC patients were included in the study. TRAIL and TRAIL receptor expression profiles were determined by immunohistochemistry. Annexin V binding revealed the apoptotic index in pancreas. Lastly, the tumor grade, tumor stage, tumor diameter, perineural invasion, and number of lymph node metastasis were used for comparison purposes. RESULTS TRAIL decoy receptor 2 (DcR2) and death receptor 4 expression were up-regulated in PDAC patients compared with noncancer patients, and the ductal cells of PDAC patients displayed significant levels of apoptosis. In addition, acinar cells from PDAC patients had higher DcR2 expression but lower death receptor 4 expression. Increased DcR2 expression was also observed in Langerhans islets of PDAC patients. CONCLUSIONS Differential alteration of TRAIL and TRAIL receptor expression profiles in PDAC patients suggest that the TRAIL/TRAIL receptor system may play a pivotal role during pancreatic carcinoma development.
Collapse
Affiliation(s)
- Ahter Dilsad Sanlioglu
- Department of Medical Biology and Genetics, Faculty of Medicine, Human Gene Therapy Unit, Akdeniz University, Antalya, Turkey.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Triggering of tumour cell apoptosis is the foundation of many cancer therapies. Death receptors of the tumour necrosis factor (TNF) superfamily have been largely characterized, as have the signals that are generated when these receptors are activated. TNF-related apoptosis-inducing ligand (TRAIL) receptors (TRAILR1 and TRAILR2) are promising targets for cancer therapy. Herein we review what is known about the molecular control of TRAIL-mediated apoptosis, the role of TRAIL in carcinogenesis and the potential therapeutic utility of recombinant TRAIL and agonistic antibodies against TRAILR1 and TRAILR2.
Collapse
Affiliation(s)
- Ricky W Johnstone
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia.
| | | | | |
Collapse
|
35
|
Sanlioglu AD, Griffith TS, Omer A, Dirice E, Sari R, Altunbas HA, Balci MK, Sanlioglu S. Molecular mechanisms of death ligand-mediated immune modulation: a gene therapy model to prolong islet survival in type 1 diabetes. J Cell Biochem 2008; 104:710-20. [PMID: 18247339 DOI: 10.1002/jcb.21677] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Type 1 diabetes results from the T cell-mediated destruction of pancreatic beta cells. Islet transplantation has recently become a potential therapeutic approach for patients with type 1 diabetes. However, islet-graft failure appears to be a challenging issue to overcome. Thus, complementary gene therapy strategies are needed to improve the islet-graft survival following transplantation. Immune modulation through gene therapy represents a novel way of attacking cytotoxic T cells targeting pancreatic islets. Various death ligands of the TNF family such as FasL, TNF, and TNF-Related Apoptosis-Inducing Ligand (TRAIL) have been studied for this purpose. The over-expression of TNF or FasL in pancreatic islets exacerbates the onset of type 1 diabetes generating lymphocyte infiltrates responsible for the inflammation. Conversely, the lack of TRAIL expression results in higher degree of islet inflammation in the pancreas. In addition, blocking of TRAIL function using soluble TRAIL receptors facilitates the onset of diabetes. These results suggested that contrary to what was observed with TNF or FasL, adenovirus mediated TRAIL gene delivery into pancreatic islets is expected to be therapeutically beneficial in the setting of experimental models of type 1 diabetes. In conclusion; this study mainly reveals the fundamental principles of death ligand-mediated immune evasion in diabetes mellitus.
Collapse
Affiliation(s)
- Ahter Dilsad Sanlioglu
- Human Gene Therapy Unit and the Department of Medical Biology and Genetics, Akdeniz University, Faculty of Medicine, 07070 Antalya, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Granci V, Bibeau F, Kramar A, Boissière-Michot F, Thézénas S, Thirion A, Gongora C, Martineau P, Del Rio M, Ychou M. Prognostic significance of TRAIL-R1 and TRAIL-R3 expression in metastatic colorectal carcinomas. Eur J Cancer 2008; 44:2312-8. [PMID: 18755584 DOI: 10.1016/j.ejca.2008.06.042] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Revised: 06/17/2008] [Accepted: 06/23/2008] [Indexed: 11/29/2022]
Abstract
Drug resistance is believed to cause treatment failure in patients with metastatic colorectal carcinoma (CRC). Resistance to chemotherapy can involve different processes, including apoptosis, whose extrinsic pathway is regulated by expression of death-inducing TRAIL-R1 and -R2 and inhibitory TRAIL-R3 and -R4 cell surface receptors. Therefore, we investigated whether variations in their expression could influence the response to 5-Fluorouracil (5-FU) in metastatic CRC. We analysed TRAIL-R 1, -2, -3 and -4 expression by immuno-histochemistry in CRC, using tissue micro arrays, and found that concomitant low/medium TRAIL-R1 and high TRAIL-R3 expression in primary CRC is significantly associated with a poor response to 5-FU-based first-line chemotherapy and with shorter progression-free survival. Specifically, the median progression-free survival was 3.1 months (poor prognostic group) versus 10.1 in the good prognostic group. Thus, the combination of TRAIL-R1 and TRAIL-R3 expression might represent a predictive and prognostic factor of the response to 5-FU-based first-line chemotherapy in patients with metastatic CRC.
Collapse
Affiliation(s)
- Virginie Granci
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, U896, Université Montpellier1, CRLC Val d'Aurelle Paul Lamarque, 208 rue des Apothicaires, F-34298 Montpellier Cedex 5, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Sanlioglu AD, Dirice E, Elpek O, Korcum AF, Balci MK, Omer A, Griffith TS, Sanlioglu S. High levels of endogenous tumor necrosis factor-related apoptosis-inducing ligand expression correlate with increased cell death in human pancreas. Pancreas 2008; 36:385-393. [PMID: 18437085 DOI: 10.1097/mpa.0b013e318158a4e5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Type 1 diabetes (T1D) has been characterized by the T cell-mediated destruction of pancreatic beta cells. Although various members of the tumor necrosis factor (TNF) family, such as Fas ligand or TNF, have recently been implicated in the development of T1D, the lack of TNF-related apoptosis-inducing ligand (TRAIL) expression or function facilitates the onset of T1D. Thus, the goal of the present study was to investigate the expression profiles of TRAIL and its receptors in human pancreas. METHODS Pancreata of 31 patients were analyzed by immunohistochemistry using antibodies developed against TRAIL and its receptors. Apoptosis was confirmed by Annexin V-fluorescein isothiocyanate binding and terminal deoxynucleotidyl transferase-mediated 2'-deoxyuridine 5'-triphosphate nick end labeling assays. RESULTS Acinar cells displayed high levels of TRAIL and death receptor 4, but only low levels of death receptor 5. In contrast, only TRAIL and TRAIL decoy receptors (DcR1, DcR2) were detected in ductal cells. Similarly, Langerhans islets expressed only TRAIL and TRAIL decoy receptor. High levels of TRAIL expression in pancreas correlated with increased number of apoptotic cells. CONCLUSIONS Although the expression of TRAIL decoy receptors might be necessary for defense from TRAIL-induced apoptosis, high levels of TRAIL may provide protection for Langerhans islets from the immunological attack of cytotoxic T cells.
Collapse
Affiliation(s)
- Ahter Dilsad Sanlioglu
- Human Gene Therapy Unit, Departments of Medical Biology and Genetics, Akdeniz University, Faculty of Medicine, Antalya, Turkey.
| | | | | | | | | | | | | | | |
Collapse
|