1
|
Wang S, Zhang M, Li T, Chen X, Wu Q, Tian D, Granot Z, Xu H, Hao J, Zhang H. A comprehensively prognostic and immunological analysis of PARP11 in pan-cancer. J Leukoc Biol 2024; 117:qiae030. [PMID: 38334307 DOI: 10.1093/jleuko/qiae030] [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: 12/12/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 02/10/2024] Open
Abstract
Poly (ADP-ribose) polymerase family member 11 (PARP11) has important immune regulatory functions in viral infection and tumor immune response. Particularly, PARP11 showed protumor activities in multiple preclinical murine models. However, no systematic pan-cancer analysis has been conducted to explore PARP11 function. In this study, we used multiple databases to assess PARP11 expression, which is associated with clinical outcomes, immune checkpoint factors, prognostic significance, genomic characteristics, and immunological aspects. The analysis revealed varying expression levels of PARP11 across different cancer types and a significant correlation between its expression and immune cell infiltration. Insights from the CellMiner database suggest a strong link between PARP11 expression and sensitivity to anticancer drugs, highlighting its potential as a therapeutic target. Moreover, PARP11 expression correlates with patient survival during anti-PD1 and anti-CTLA4 treatments, suggesting that PARP11 would be a predictor of immune checkpoint inhibitor treatment. In summary, PARP11 would be a potential immunoregulatory target and a diagnosis and prognosis marker for certain types of cancers. The detailed mechanisms of PARP11 in tumor immune responses need to be further investigated.
Collapse
Affiliation(s)
- Shengli Wang
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, 79 Kangning Rd, Zhuhai, 519000, Guangdong, P.R. China
- Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, 601 W Huangpu Ave, Guangzhou, 510632, Guangdong, P.R. China
| | - Mingyue Zhang
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, 79 Kangning Rd, Zhuhai, 519000, Guangdong, P.R. China
- Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, 601 W Huangpu Ave, Guangzhou, 510632, Guangdong, P.R. China
| | - Tao Li
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, 79 Kangning Rd, Zhuhai, 519000, Guangdong, P.R. China
- Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, 601 W Huangpu Ave, Guangzhou, 510632, Guangdong, P.R. China
| | - Xinru Chen
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, 79 Kangning Rd, Zhuhai, 519000, Guangdong, P.R. China
- Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, 601 W Huangpu Ave, Guangzhou, 510632, Guangdong, P.R. China
| | - Qinhan Wu
- College of Life Sciences, Nankai University, 94 Weijin Rd, Tianjin, 300071, P.R. China
| | - Dan Tian
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshaner St, Guangzhou, 510080, Guangdong, P.R. China
| | - Zvi Granot
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University Hadassah Medical School, Ein Kerem, 9112102, Jerusalem, Israel
| | - Hongbiao Xu
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshaner St, Guangzhou, 510275, Guangdong, P.R. China
| | - Jianlei Hao
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, 79 Kangning Rd, Zhuhai, 519000, Guangdong, P.R. China
- Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, 601 W Huangpu Ave, Guangzhou, 510632, Guangdong, P.R. China
| | - Hongru Zhang
- College of Life Sciences, Nankai University, 94 Weijin Rd, Tianjin, 300071, P.R. China
| |
Collapse
|
2
|
Zhang DH, Liang C, Hu SY, Huang XY, Yu L, Meng XL, Guo XJ, Zeng HY, Chen Z, Zhang L, Pei YZ, Ye M, Cai JB, Huang PX, Shi YH, Ke AW, Chen Y, Ji Y, Shi YG, Zhou J, Fan J, Yang GH, Sun QM, Shi GM, Lu JC. Application of a single-cell-RNA-based biological-inspired graph neural network in diagnosis of primary liver tumors. J Transl Med 2024; 22:883. [PMID: 39354613 PMCID: PMC11445937 DOI: 10.1186/s12967-024-05670-1] [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: 04/14/2024] [Accepted: 09/12/2024] [Indexed: 10/03/2024] Open
Abstract
Single-cell technology depicts integrated tumor profiles including both tumor cells and tumor microenvironments, which theoretically enables more robust diagnosis than traditional diagnostic standards based on only pathology. However, the inherent challenges of single-cell RNA sequencing (scRNA-seq) data, such as high dimensionality, low signal-to-noise ratio (SNR), sparse and non-Euclidean nature, pose significant obstacles for traditional diagnostic approaches. The diagnostic value of single-cell technology has been largely unexplored despite the potential advantages. Here, we present a graph neural network-based framework tailored for molecular diagnosis of primary liver tumors using scRNA-seq data. Our approach capitalizes on the biological plausibility inherent in the intercellular communication networks within tumor samples. By integrating pathway activation features within cell clusters and modeling unidirectional inter-cellular communication, we achieve robust discrimination between malignant tumors (including hepatocellular carcinoma, HCC, and intrahepatic cholangiocarcinoma, iCCA) and benign tumors (focal nodular hyperplasia, FNH) by scRNA data of all tissue cells and immunocytes only. The efficacy to distinguish iCCA from HCC was further validated on public datasets. Through extending the application of high-throughput scRNA-seq data into diagnosis approaches focusing on integrated tumor microenvironment profiles rather than a few tumor markers, this framework also sheds light on minimal-invasive diagnostic methods based on migrating/circulating immunocytes.
Collapse
Affiliation(s)
- Dao-Han Zhang
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chen Liang
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shu-Yang Hu
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiao-Yong Huang
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Liver Cancer Institute, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education of the People's Republic of China, Shanghai, 200032, China
| | - Lei Yu
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Liver Cancer Institute, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education of the People's Republic of China, Shanghai, 200032, China
| | - Xian-Long Meng
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Liver Cancer Institute, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education of the People's Republic of China, Shanghai, 200032, China
| | - Xiao-Jun Guo
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Liver Cancer Institute, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education of the People's Republic of China, Shanghai, 200032, China
| | - Hai-Ying Zeng
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhen Chen
- Clinical Research Unit, Institute of Clinical Science, Zhongshan Hospital of Fudan University, Shanghai, 200032, China
| | - Lv Zhang
- Clinical Research Unit, Institute of Clinical Science, Zhongshan Hospital of Fudan University, Shanghai, 200032, China
| | - Yan-Zi Pei
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Mu Ye
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jia-Bin Cai
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Pei-Xin Huang
- Liver Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Ying-Hong Shi
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Liver Cancer Institute, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education of the People's Republic of China, Shanghai, 200032, China
| | - Ai-Wu Ke
- Liver Cancer Institute, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education of the People's Republic of China, Shanghai, 200032, China
| | - Yi Chen
- Liver Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yujiang Geno Shi
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Liver Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Liver Cancer Institute, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education of the People's Republic of China, Shanghai, 200032, China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Liver Cancer Institute, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education of the People's Republic of China, Shanghai, 200032, China
- Department of Liver Surgery, Shanghai Geriatric Medical Center, Fudan University, Shanghai, 200032, China
| | - Guo-Huan Yang
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Qi-Man Sun
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Guo-Ming Shi
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Liver Cancer Institute, Fudan University, Shanghai, 200032, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education of the People's Republic of China, Shanghai, 200032, China.
- Clinical Research Unit, Institute of Clinical Science, Zhongshan Hospital of Fudan University, Shanghai, 200032, China.
- Department of Liver Surgery, Shanghai Geriatric Medical Center, Fudan University, Shanghai, 200032, China.
| | - Jia-Cheng Lu
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Liver Cancer Institute, Fudan University, Shanghai, 200032, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education of the People's Republic of China, Shanghai, 200032, China.
| |
Collapse
|
3
|
Švajger U, Kamenšek U. Interleukins and interferons in mesenchymal stromal stem cell-based gene therapy of cancer. Cytokine Growth Factor Rev 2024; 77:76-90. [PMID: 38508954 DOI: 10.1016/j.cytogfr.2024.03.002] [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: 02/05/2024] [Revised: 03/07/2024] [Accepted: 03/11/2024] [Indexed: 03/22/2024]
Abstract
The tumor microenvironment is importantly shaped by various cytokines, where interleukins (ILs) and interferons (IFNs) shape the balance of immune activity within tumor niche and associated lymphoid organs. Their importance in activation and tuning of both innate and adaptive immune responses prompted their use in several clinical trials, albeit with limited therapeutic efficacy and risk of toxicity due to systemic administration. Increasing preclinical evidence suggests that local delivery of ILs and IFNs could significantly increase their effectiveness, while simultaneously attenuate the known side effects and issues related to their biological activity. A prominent way to achieve this is to use cell-based delivery vehicles. For this purpose, mesenchymal stromal stem cells (MSCs) are considered an almost ideal candidate. Namely, MSCs can be obtained in large quantities and from obtainable sources (e.g. umbilical cord or adipose tissue), their ex vivo expansion is relatively straightforward compared to other cell types and they possess very low immunogenicity making them suitable for allogeneic use. Importantly, MSCs have shown an intrinsic capacity to respond to tumor-directed chemotaxis. This review provides a focused and detailed discussion on MSC-based gene therapy using ILs and IFNs, engineering techniques and insights on potential future advancements.
Collapse
Affiliation(s)
- Urban Švajger
- Slovenian Institute for Transfusion Medicine, Department for Therapeutic Services, Šlajmerjeva Ulica 6, Ljubljana SI-1000, Slovenia; Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, Ljubljana SI-1000, Slovenia.
| | - Urška Kamenšek
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloška Cesta 2, Ljubljana SI-1000, Slovenia; Biotechnical Faculty, University of Ljubljana, Jamnikarjeva Ulica 101, Ljubljana SI-1000, Slovenia
| |
Collapse
|
4
|
Wang X, Wang L, Liu W, Liu X, Jia X, Feng X, Li F, Zhu R, Yu J, Zhang H, Wu H, Wu J, Wang C, Yu B, Yu X. Dose-related immunomodulatory effects of recombinant TRAIL in the tumor immune microenvironment. J Exp Clin Cancer Res 2023; 42:216. [PMID: 37605148 PMCID: PMC10464183 DOI: 10.1186/s13046-023-02795-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/10/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND In addition to specifically inducing tumor cell apoptosis, recombinant tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) has also been reported to influence the cancer immune microenvironment; however, its underlying effects and mechanisms remain unclear. Investigating the immunomodulatory effects and mechanisms of recombinant TRAIL in the tumor microenvironment (TME) may provide an important perspective and facilitate the exploration of novel TRAIL strategies for tumor therapy. METHODS Immunocompetent mice with different tumors were treated with three doses of recombinant TRAIL, and then the tumors were collected for immunological detection and mechanistic investigation. Methodological approaches include flow cytometry analysis and single-cell sequencing. RESULTS In an immunocompetent mouse model, recombinant soluble mouse TRAIL (smTRAIL) had dose-related immunomodulatory effects. The optimal dose of smTRAIL (2 mg/kg) activated innate immune cells and CD8+ T cells, whereas higher doses of smTRAIL (8 mg/kg) promoted the formation of a tumor-promoting immune microenvironment to counteract the apoptotic effects on tumor cells. The higher doses of smTRAIL treatment promoted M2-like macrophage recruitment and polarization and increased the production of protumor inflammatory cytokines, such as IL-10, which deepened the suppression of natural killer (NK) cells and CD8+ T cells in the tumor microenvironment. By constructing an HU-HSC-NPG.GM3 humanized immune system mouse model, we further verified the immunomodulatory effects induced by recombinant soluble human TRAIL (shTRAIL) and found that combinational administration of shTRAIL and trabectedin, a macrophage-targeting drug, could remodel the tumor immune microenvironment, further enhance antitumor immunity, and strikingly improve antitumor effects. CONCLUSION Our results highlight the immunomodulatory role of recombinant TRAIL and suggest promising therapeutic strategies for clinical application.
Collapse
Affiliation(s)
- Xupu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Lizheng Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
- Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, and the Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | - Wenmo Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Xinyao Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Xinyuan Jia
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Xinyao Feng
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Fangshen Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Rui Zhu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Jiahao Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Haihong Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Hui Wu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Jiaxin Wu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Chu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Bin Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China.
| | - Xianghui Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China.
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, China.
| |
Collapse
|
5
|
Higuchi S, Yoshida S, Minematsu T, Hatano Y, Notsu A, Ichinose T. Comparison of the Effects of Bathing and the Dry Technique on the Skin Condition of Early Neonates: A Prospective Observational Study. Ann Dermatol 2023; 35:256-265. [PMID: 37550226 PMCID: PMC10407340 DOI: 10.5021/ad.22.098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 03/24/2023] [Accepted: 05/02/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND In Japan, neonates have typically been bathed in a bathtub immediately after birth because bathing is a custom for cleansing impurities. However, dry technique has been introduced into many institutions since 2000. There is little scientific evidence on the benefit or harmfulness of either method to neonatal skin, and consequently, opinion remains split on which method is superior. OBJECTIVE The purpose of the present study was to determine whether bathing or the dry technique of cleaning is better in maintaining skin health in the early neonatal period. METHODS Transepidermal water loss (TEWL) and skin pH, considered an index of skin barrier function, were measured in each group. Tumor necrosis factor (TNF)-alpha and interleukin (IL)-6, which are inflammatory cytokines released by keratinocytes, were measured by skin blotting. RESULTS TEWL and skin pH of neonates were lower with the dry technique than with bathing. The expression level of IL-6 and TNF-α in chest skin of neonates was higher with bathing than with the dry technique. CONCLUSION These results suggest that the dry technique may maintain skin health better than bathing in the early neonatal period.
Collapse
Affiliation(s)
- Sachi Higuchi
- Department of Midwifery, Oita University of Nursing and Health Sciences, Oita, Japan.
| | - Seiichi Yoshida
- Department of Health Sciences, Oita University of Nursing and Health Sciences, Oita, Japan
| | - Takeo Minematsu
- Department of Adult Nursing, Ishikawa Prefectural Nursing University, Ishikawa, Japan
| | - Yutaka Hatano
- Department of Dermatology, Faculty of Medicine, Oita University, Oita, Japan
| | - Akifumi Notsu
- Clinical Research Promotion Unit, Shizuoka Cancer Center, Shizuoka, Japan
| | | |
Collapse
|
6
|
Pimentel JM, Zhou JY, Wu GS. The Role of TRAIL in Apoptosis and Immunosurveillance in Cancer. Cancers (Basel) 2023; 15:2752. [PMID: 37345089 DOI: 10.3390/cancers15102752] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/01/2023] [Accepted: 05/10/2023] [Indexed: 06/23/2023] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily that selectively induces apoptosis in tumor cells without harming normal cells, making it an attractive agent for cancer therapy. TRAIL induces apoptosis by binding to and activating its death receptors DR4 and DR5. Several TRAIL-based treatments have been developed, including recombinant forms of TRAIL and its death receptor agonist antibodies, but the efficacy of TRAIL-based therapies in clinical trials is modest. In addition to inducing cancer cell apoptosis, TRAIL is expressed in immune cells and plays a critical role in tumor surveillance. Emerging evidence indicates that the TRAIL pathway may interact with immune checkpoint proteins, including programmed death-ligand 1 (PD-L1), to modulate PD-L1-based tumor immunotherapies. Therefore, understanding the interaction between TRAIL and the immune checkpoint PD-L1 will lead to the development of new strategies to improve TRAIL- and PD-L1-based therapies. This review discusses recent findings on TRAIL-based therapy, resistance, and its involvement in tumor immunosurveillance.
Collapse
Affiliation(s)
- Julio M Pimentel
- Molecular Therapeutics Program, Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Cancer Biology Program, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Department of Oncology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Jun-Ying Zhou
- Molecular Therapeutics Program, Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Department of Oncology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Gen Sheng Wu
- Molecular Therapeutics Program, Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Cancer Biology Program, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Department of Oncology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Department of Pathology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
7
|
Zhang W, Yin Q, Huang H, Lu J, Qin H, Chen S, Zhang W, Su X, Sun W, Dong Y, Li Q. Personal Neoantigens From Patients With NSCLC Induce Efficient Antitumor Responses. Front Oncol 2021; 11:628456. [PMID: 33928024 PMCID: PMC8076796 DOI: 10.3389/fonc.2021.628456] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/23/2021] [Indexed: 12/26/2022] Open
Abstract
Objective To develop a neoantigen-targeted personalized cancer treatment for non-small cell lung cancer (NSCLC), neoantigens were obtained from collected human lung cancer samples, and the utility of neoantigen and neoantigen-reactive T cells (NRTs) was assessed. Methods Tumor specimens from three patients with NSCLC were obtained and analyzed by whole-exome sequencing, and neoantigens were predicted accordingly. Dendritic cells and T lymphocytes were isolated, NRTs were elicited and IFN-γ ELISPOT tests were conducted. HLA-A2.1/Kb transgenic mice were immunized with peptides from HLA-A*02:01+patient with high immunogenicity, and NRTs were subjected to IFN-γ, IL-2 and TNF-α ELISPOT as well as time-resolved fluorescence assay for cytotoxicity assays to verify the immunogenicity in vitro. The HLA-A*02:01+lung cancer cell line was transfected with minigene and inoculated into the flanks of C57BL/6nu/nu mice and the NRTs induced by the immunogenic polypeptides from autologous HLA-A2.1/Kb transgenic mice were adoptively transfused to verify their immunogenicity in vivo. Results Multiple putative mutation-associated neoantigens with strong affinity for HLA were selected from each patient. Immunogenic neoantigen were identified in all three NSCLC patients, the potency of ACAD8-T105I, BCAR1-G23V and PLCG1-M425L as effective neoantigen to active T cells in suppressing tumor growth was further proven both in vitro and in vivo using HLA-A2.1/Kb transgenic mice and tumor-bearing mouse models. Conclusion Neoantigens with strong immunogenicity can be screened from NSCLC patients through the whole-exome sequencing of patient specimens and machine-learning-based neoantigen predictions. NRTs shown efficient antitumor responses in transgenic mice and tumor-bearing mouse models. Our results indicate that the development of neoantigen-based personalized immunotherapies in NSCLC is possible. Precis Neoantigens with strong immunogenicity were screened from NSCLC patients. This research provides evidence suggesting that neoantigen-based therapy might serve as feasible treatment for NSCLC.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Qi Yin
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Haidong Huang
- Department of Pulmonary and Critical Care Medicine, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jingjing Lu
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Hao Qin
- Department of Pulmonary and Critical Care Medicine, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Si Chen
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Wenjun Zhang
- Department of Emergency, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiaoping Su
- School of Basic Medicine, Wenzhou Medical University, Wenzhou Tea Mountain Higher Education Park, Wenzhou, China
| | - Weihong Sun
- Biotherapy Center, Qingdao Central Hospital, The Second Affiliated Hospital, Qingdao University, Qingdao, China
| | - Yuchao Dong
- Department of Pulmonary and Critical Care Medicine, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Qiang Li
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| |
Collapse
|
8
|
Karimdadi Sariani O, Eghbalpour S, Kazemi E, Rafiei Buzhani K, Zaker F. Pathogenic and therapeutic roles of cytokines in acute myeloid leukemia. Cytokine 2021; 142:155508. [PMID: 33810945 DOI: 10.1016/j.cyto.2021.155508] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/14/2021] [Accepted: 03/15/2021] [Indexed: 02/07/2023]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease with high mortality that accounts for the most common acute leukemia in adults. Despite all progress in the therapeutic strategies and increased rate of complete remission, many patients will eventually relapse and die from the disease. Cytokines as molecular messengers play a pivotal role in the immune system. The imbalance release of cytokine has been shown to exert a significant influence on the progression of hematopoietic malignancies including acute myeloid leukemia. This article aimed to summarize current knowledge about cytokines and their critical roles in the pathogenesis, treatment, and survival of AML patients.
Collapse
Affiliation(s)
- Omid Karimdadi Sariani
- Department of Genetics, College of Science, Islamic Azad University, Kazerun Branch, Kazerun, Iran
| | - Sara Eghbalpour
- School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Elahe Kazemi
- Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Farhad Zaker
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Yu Y, Zhang J, Ni L, Zhu Y, Yu H, Teng Y, Lin L, Xue Z, Xue X, Shen X, Song H, Su X, Sun W, Cai Z. Neoantigen-reactive T cells exhibit effective anti-tumor activity against colorectal cancer. Hum Vaccin Immunother 2021; 18:1-11. [PMID: 33689574 PMCID: PMC8920255 DOI: 10.1080/21645515.2021.1891814] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Neoantigens play a crucial role in cancer immunotherapy. However, the effectiveness and safety of neoantigen-based immunotherapies in patients with colorectal cancer (CRC), particularly in the Chinese population, have not been well studied. This study explored the feasibility and effectiveness of neoantigens in the treatment of CRC. Whole-exome sequencing (WES) and transcriptome sequencing were used to identify somatic mutations, RNA expression, and human leukocyte antigen (HLA) alleles. Neoantigen candidates were predicted, and immunogenicity was assessed. The neoantigens TSHZ3-L523P, RARA-R83H, TP53-R248W, EYA2-V333I, and NRAS-G12D from Patient 4 (PW4); TASP1-P161L, RAP1GAP-S215R, MOSPD1-V63I, and NAV2-D1973N from Patient 10 (PW10); and HAVCR2-F39V, SEC11A-R11L, SMPDL3B-T452M, LRFN3-R118Q, and ULK1-S248L from Patient 11 (HLA-A0201+PW11) induced a heightened neoantigen-reactive T cell (NRT) response as compared with the controls in peripheral blood lymphocytes (PBLs) isolated from patients with CRC. In addition, we identified neoantigen-containing peptides SEC11A-R11L and ULK1-S248L from HLA-A0201+PW11, which more effectively elicited specific CTL responses than the corresponding native peptides in PBLs isolated from HLA-A0201+PW11 as well as in HLA-A2.1/Kb transgenic mice. Importantly, adoptive transfer of NRTs induced by vaccination with two mutant peptides could effectively inhibit tumor growth in tumor-bearing mouse models. These data indicate that neoantigen-containing peptides with high immunogenicity represent promising candidates for peptide-mediated personalized therapy. Abbreviations: CRC: colorectal cancer; DCs: dendritic cells; ELISPOT: enzyme-linked immunosorbent spot; E:T: effector:target; HLA: human leukocyte antigen; MHC: major histocompatibility complex; Mut: mutant type; NGS: next-generation sequencing; NRTs: neoantigen-reactive T cells; PBMCs: peripheral blood mononuclear cells; STR: short tandem repeat; PBLs: peripheral blood lymphocytes; PBS: phosphate-buffered saline; PD-1: programmed cell death protein 1; TILs: tumor-infiltrating lymphocytes; RNA-seq: RNA sequencing; Tg: transgenic; TMGs: tandem minigenes; WES: whole-exome sequencing; WT: wild-type.
Collapse
Affiliation(s)
- Yaojun Yu
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jing Zhang
- Department of Gastroenterology, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Leyi Ni
- Department of Gastroenterology, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuesheng Zhu
- Department of Gastroenterology, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hejie Yu
- Department of Gastroenterology, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yangyang Teng
- Department of Gastroenterology, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Limiao Lin
- Department of Gastroenterology, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhanxiong Xue
- Department of Gastroenterology, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiangyang Xue
- Department of Oncology, Wenzhou Medical University School of Basic Medicine, Wenzhou, Zhejiang, China
| | - Xian Shen
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haiping Song
- Department of Oncology, Qingdao Central Hospital, The Second Affiliated Hospital, Qingdao University, Qingdao, China
| | - Xiaoping Su
- Department of Oncology, Wenzhou Medical University School of Basic Medicine, Wenzhou, Zhejiang, China
| | - Weihong Sun
- Department of Oncology, Biotherapy Center, Qingdao Central Hospital, The Second Affiliated Hospital, Qingdao University, Qingdao, China
| | - Zhenzhai Cai
- Department of Gastroenterology, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
10
|
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
|
11
|
Wang Y, Luo M, Chen Y, Wang Y, Zhang B, Ren Z, Bao L, Wang Y, Wang JE, Fu YX, Luo W, Wang Y. ZMYND8 Expression in Breast Cancer Cells Blocks T-Lymphocyte Surveillance to Promote Tumor Growth. Cancer Res 2020; 81:174-186. [PMID: 33148660 DOI: 10.1158/0008-5472.can-20-1710] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 10/05/2020] [Accepted: 10/30/2020] [Indexed: 11/16/2022]
Abstract
Emerging studies indicate that DNA damage in cancer cells triggers antitumor immunity, but its intrinsic regulatory mechanism in breast cancer cells remains poorly understood. Here, we show that ZMYND8 is upregulated and inhibits micronucleus formation and DNA damage in breast cancer cells. Loss of ZMYND8 triggered activation of the DNA sensor cyclic guanosine monophosphate-adenosine monophosphate synthase in micronuclei, leading to further activation of the downstream signaling effectors stimulator of IFN genes and NF-κB, but not TANK-binding kinase 1 and IFN regulatory factor 3, thereby inducing the expression of IFNβ and IFN-stimulated genes (ISG) in breast cancer cells in vitro and tumors in vivo. ZMYND8 knockout (KO) in breast cancer cells promoted infiltration of CD4+ and CD8+ T cells, leading to tumor inhibition in syngeneic mouse models, which was significantly attenuated by treatment of anti-CD4/CD8-depleting antibodies or anti-IFNAR1 antibody and in immunodeficient Rag1 KO mice. In human breast tumors, ZMYND8 was negatively correlated with ISGs, CD4, CD8A, CD8B, and the tumor-lymphocyte infiltration phenotype. Collectively, these findings demonstrate that maintenance of genome stability by ZMYND8 causes breast cancer cells to evade cytotoxic T-lymphocyte surveillance, which leads to tumor growth. SIGNIFICANCE: These findings show that ZMYND8 is a new negative and intrinsic regulator of the innate immune response in breast tumor cells, and ZMYND8 may be a possible target for antitumor immunotherapy.
Collapse
Affiliation(s)
- Yong Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Maowu Luo
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Yan Chen
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Yijie Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Bo Zhang
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Zhenhua Ren
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Lei Bao
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Yanan Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Jennifer E Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Yang-Xin Fu
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Weibo Luo
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas. .,Department of Pharmacology, UT Southwestern Medical Center, Dallas, Texas
| | - Yingfei Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas. .,Department of Neurology, UT Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
12
|
Sag D, Ayyildiz ZO, Gunalp S, Wingender G. The Role of TRAIL/DRs in the Modulation of Immune Cells and Responses. Cancers (Basel) 2019; 11:cancers11101469. [PMID: 31574961 PMCID: PMC6826877 DOI: 10.3390/cancers11101469] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/09/2019] [Accepted: 09/20/2019] [Indexed: 12/26/2022] Open
Abstract
Expression of TRAIL (tumor necrosis factor–related apoptosis–inducing ligand) by immune cells can lead to the induction of apoptosis in tumor cells. However, it becomes increasingly clear that the interaction of TRAIL and its death receptors (DRs) can also directly impact immune cells and influence immune responses. Here, we review what is known about the role of TRAIL/DRs in immune cells and immune responses in general and in the tumor microenvironment in particular.
Collapse
Affiliation(s)
- Duygu Sag
- Izmir Biomedicine and Genome Center (IBG), 35340 Balcova/Izmir, Turkey.
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| | - Zeynep Ozge Ayyildiz
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| | - Sinem Gunalp
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| | - Gerhard Wingender
- Izmir Biomedicine and Genome Center (IBG), 35340 Balcova/Izmir, Turkey.
- Department of Biomedicine and Health Technologies, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| |
Collapse
|
13
|
Castiello L, Aricò E, D'Agostino G, Santodonato L, Belardelli F. In situ Vaccination by Direct Dendritic Cell Inoculation: The Coming of Age of an Old Idea? Front Immunol 2019; 10:2303. [PMID: 31611878 PMCID: PMC6773832 DOI: 10.3389/fimmu.2019.02303] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/11/2019] [Indexed: 12/18/2022] Open
Abstract
For more than 25 years, dendritic cell (DC) based vaccination has flashily held promises to represent a therapeutic approach for cancer treatment. While the vast majority of studies has focused on the use of antigen loaded DC, the intratumoral delivery of unloaded DC aiming at in situ vaccination has gained much less attention. Such approach grounds on the ability of inoculated DC to internalize and process antigens directly released by tumor (usually in combination with cell-death-inducing agents) to activate broad patient-specific antitumor T cell response. In this review, we highlight the recent studies in both solid and hematological tumors showing promising clinical results and discuss the main pitfalls and advantages of this approach for endogenous cancer vaccination. Lastly, we discuss how in situ vaccination by DC inoculation may fit with current immunotherapy approaches to expand and prolong patient response.
Collapse
Affiliation(s)
- Luciano Castiello
- FaBioCell, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Eleonora Aricò
- FaBioCell, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | | | - Laura Santodonato
- FaBioCell, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Filippo Belardelli
- Consiglio Nazionale Delle Ricerche, Institute of Translational Pharmacology, Rome, Italy
| |
Collapse
|
14
|
Paim AC, Cummins NW, Natesampillai S, Garcia-Rivera E, Kogan N, Neogi U, Sönnerborg A, Sperk M, Bren GD, Deeks S, Polley E, Badley AD. HIV elite control is associated with reduced TRAILshort expression. AIDS 2019; 33:1757-1763. [PMID: 31149947 PMCID: PMC6873462 DOI: 10.1097/qad.0000000000002279] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) dependent apoptosis has been implicated in CD4 T-cell death and immunologic control of HIV-1 infection. We have described a splice variant called TRAILshort, which is a dominant negative ligand that antagonizes TRAIL-induced cell death in the context of HIV-1 infection. HIV-1 elite controllers naturally control viral replication for largely unknown reasons. Since enhanced death of infected cells might be responsible, as might occur in situations of low (or inhibited) TRAILshort, we tested whether there was an association between elite controller status and reduced levels of TRAILshort expression. DESIGN Cohort study comparing TRAILshort and full length TRAIL expression between HIV-1 elite controllers and viremic progressors from two independent populations. METHODS TRAILshort and TRAIL gene expression in peripheral blood mononuclear cells (PBMCs) was determined by RNA-seq. TRAILshort and TRAIL protein expression in plasma was determined by antibody bead array and proximity extension assay respectively. RESULTS HIV-1 elite controllers expressed less TRAILshort transcripts in PBMCs (P = 0.002) and less TRAILshort protein in plasma (P < 0.001) than viremic progressors. CONCLUSION Reduced TRAILshort expression in PBMCs and plasma is associated with HIV-1 elite controller status.
Collapse
Affiliation(s)
- Ana C Paim
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota
| | - Nathan W Cummins
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota
| | | | | | | | - Ujjwal Neogi
- Division of Clinical Microbiology, Karolinska Institutet, Stockholm, Sweden
| | - Anders Sönnerborg
- Division of Clinical Microbiology, Karolinska Institutet, Stockholm, Sweden
| | - Maike Sperk
- Division of Clinical Microbiology, Karolinska Institutet, Stockholm, Sweden
| | - Gary D Bren
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota
| | - Steve Deeks
- Division of Infectious Diseases, University of California, San Francisco, San Francisco, California
| | - Eric Polley
- Division of Biomedical Statistics and Informatics
| | - Andrew D Badley
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
15
|
Natesampillai S, Paim AC, Cummins NW, Chandrasekar AP, Bren GD, Lewin SR, Kiem HP, Badley AD. TRAILshort Protects against CD4 T Cell Death during Acute HIV Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:718-724. [PMID: 31189571 PMCID: PMC6785036 DOI: 10.4049/jimmunol.1900271] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/21/2019] [Indexed: 02/07/2023]
Abstract
CD4 T cells from HIV-1 infected patients die at excessive rates compared to those from uninfected patients, causing immunodeficiency. We previously identified a dominant negative ligand that antagonizes the TRAIL-dependent pathway of cell death, which we called TRAILshort. Because the TRAIL pathway has been implicated in CD4 T cell death occurring during HIV-1 infection, we used short hairpin RNA knockdown, CRISPR deletion, or Abs specific for TRAILshort to determine the effect of inhibiting TRAILshort on the outcome of experimental acute HIV infection in vitro. Strikingly, all three approaches to TRAILshort deletion/inhibition enhanced HIV-induced death of both infected and uninfected human CD4 T cells. Thus, TRAILshort impacts T cell dynamics during HIV infection, and inhibiting TRAILshort causes more HIV-infected and uninfected bystander cells to die. TRAILshort is, therefore, a host-derived, host-adaptive mechanism to limit the effects of TRAIL-induced cell death. Further studies on the effects of TRAILshort in other disease states are warranted.
Collapse
Affiliation(s)
| | - Ana C Paim
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN 55905
| | - Nathan W Cummins
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN 55905
| | | | - Gary D Bren
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN 55905
| | - Sharon R Lewin
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria 3000, Australia
- Department of Infectious Diseases, Alfred Health and Monash University, Melbourne, Victoria 3004, Australia
| | - Hans-Peter Kiem
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109; and
| | - Andrew D Badley
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN 55905;
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
16
|
Wu H, Zhuang Q, Xu J, Xu L, Zhao Y, Wang C, Yang Z, Shen F, Liu Z, Peng R. Cell-Penetrating Peptide Enhanced Antigen Presentation for Cancer Immunotherapy. Bioconjug Chem 2019; 30:2115-2126. [DOI: 10.1021/acs.bioconjchem.9b00245] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Hanfei Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Rd, Suzhou, Jiangsu 215123, P. R. China
| | - Qi Zhuang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Rd, Suzhou, Jiangsu 215123, P. R. China
| | - Jun Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Rd, Suzhou, Jiangsu 215123, P. R. China
| | - Ligeng Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Rd, Suzhou, Jiangsu 215123, P. R. China
| | - Yuhuan Zhao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Rd, Suzhou, Jiangsu 215123, P. R. China
| | - Chenya Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Rd, Suzhou, Jiangsu 215123, P. R. China
| | - Zongjin Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Rd, Suzhou, Jiangsu 215123, P. R. China
| | - Fengyun Shen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Rd, Suzhou, Jiangsu 215123, P. R. China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Rd, Suzhou, Jiangsu 215123, P. R. China
| | - Rui Peng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Rd, Suzhou, Jiangsu 215123, P. R. China
| |
Collapse
|
17
|
Importance of TRAIL Molecular Anatomy in Receptor Oligomerization and Signaling. Implications for Cancer Therapy. Cancers (Basel) 2019; 11:cancers11040444. [PMID: 30934872 PMCID: PMC6521207 DOI: 10.3390/cancers11040444] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 12/12/2022] Open
Abstract
(TNF)-related apoptosis-inducing ligand (TRAIL) is able to activate the extrinsic apoptotic pathway upon binding to DR4/TRAIL-R1 and/or DR5/TRAIL-R2 receptors. Structural data indicate that TRAIL functions as a trimer that can engage three receptor molecules simultaneously, resulting in receptor trimerization and leading to conformational changes in TRAIL receptors. However, receptor conformational changes induced by the binding of TRAIL depend on the molecular form of this death ligand, and not always properly trigger the apoptotic cascade. In fact, TRAIL exhibits a much stronger pro-apoptotic activity when is found as a transmembrane protein than when it occurs as a soluble form and this enhanced biological activity is directly linked to its ability to cluster TRAIL receptors in supra-molecular structures. In this regard, cells involved in tumor immunosurveillance, such as activated human T cells, secrete endogenous TRAIL as a transmembrane protein associated with lipid microvesicles called exosomes upon T-cell reactivation. Consequently, it seems clear that a proper oligomerization of TRAIL receptors, which leads to a strong apoptotic signaling, is crucial for inducing apoptosis in cancer cells upon TRAIL treatment. In this review, the current knowledge of oligomerization status of TRAIL receptors is discussed as well as the implications for cancer treatment when using TRAIL-based therapies.
Collapse
|
18
|
Diversity of dendritic cells generated from umbilical cord or adult peripheral blood precursors. Cent Eur J Immunol 2018; 43:306-313. [PMID: 30588175 PMCID: PMC6305608 DOI: 10.5114/ceji.2018.80050] [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: 02/08/2017] [Accepted: 04/24/2017] [Indexed: 02/01/2023] Open
Abstract
Following the discovery of methods to generate large numbers of specific dendritic cells (DCs) ex vivo, the possibility of exploiting these cells in immunotherapeutic strategies will become a reality. It seems to be rationally to analyse the influence of the precursor source for further features and applications. For the needs of the given project DCs were derived from precursors derived from adult peripheral blood (APB) and umbilical cord blood (UCB). During some expansions of UCB CD34+ cells were separated giving non-adherent DCs (NA-DCs) or adherent DCs (A-DCs), whereas DCs derived from UCB precursors without separation gave rise to All-DCs. DC subpopulations were stimulated by lipopolysaccharides (LPS) or interferon-γ (IFN-γ), and afterwards the morphology, phenotype, and stimulatory properties were analysed. Our findings demonstrated that DCs generated from APB and UCB precursors were not equivalent and exhibited opposite features when expanded in comparable conditions. Additionally, all three subpopulations of UCB-derived DCs presented functional dissimilarities. Based on our results we concluded that the precursor source and the composition of media must be considered as crucial to the success of potential therapeutic application.
Collapse
|
19
|
Tyrinova T, Leplina O, Mishinov S, Tikhonova M, Kalinovskiy A, Chernov S, Dolgova E, Stupak V, Voronina E, Bogachev S, Shevela E, Ostanin A, Chernykh E. Defective Dendritic Cell Cytotoxic Activity of High-Grade Glioma Patients' Results from the Low Expression of Membrane TNFα and Can Be Corrected In Vitro by Treatment with Recombinant IL-2 or Exogenic Double-Stranded DNA. J Interferon Cytokine Res 2018; 38:298-310. [PMID: 29932796 DOI: 10.1089/jir.2017.0084] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Besides initiation of tumor-specific T cell immunity, dendritic cells (DCs) are endowed with tumoricidal activity. Previously, we showed that monocyte-derived DCs of high-grade glioma patients generated in the presence of interferon alpha (IFNα) (IFN-DCs) have impaired cytotoxic activity against tumor necrosis factor alpha (TNFα)-sensitive HEp-2 tumor cells. Herein, we demonstrate that decreased transmembrane TNFα (tmTNFα) expression, but not soluble TNFα (sTNFα) production by high-grade glioma patient IFN-DCs, determines the defective tumoricidal activity against TNFα-sensitive HEp-2 cells. Blocking TNFα-converting enzyme or stimulation of patient IFN-DCs with rIL-2 or dsDNA enhances tmTNFα expression on IFN-DCs and significantly increases their cytotoxicity. Decreased tmTNFα expression on patient IFN-DCs is not caused by downregulation of pNFκB. Neither rIL-2 nor dsDNA upregulates tmTNFα expression on patient IFN-DCs via an increase of pNFκB. The current study shows an important role of tmTNFα as mediator of IFN-DC tumoricidal activity and as molecular target for the restoration of defective DC killer activity in high-grade glioma patients.
Collapse
Affiliation(s)
- Tamara Tyrinova
- 1 Laboratory of Cellular Immunotherapy, Institute of Fundamental and Clinical Immunology , Novosibirsk, Russia
| | - Olga Leplina
- 1 Laboratory of Cellular Immunotherapy, Institute of Fundamental and Clinical Immunology , Novosibirsk, Russia
| | - Sergey Mishinov
- 2 Department of Neurosurgery, Novosibirsk Research Institute of Traumatology and Orthopedics named after Ya.L. Zivian , Novosibirsk, Russia
| | - Marina Tikhonova
- 1 Laboratory of Cellular Immunotherapy, Institute of Fundamental and Clinical Immunology , Novosibirsk, Russia
| | - Anton Kalinovskiy
- 3 Department of Neurosurgery, Federal Neurosurgical Center , Novosibirsk, Russia
| | - Sergey Chernov
- 3 Department of Neurosurgery, Federal Neurosurgical Center , Novosibirsk, Russia
| | - Evgeniya Dolgova
- 4 Laboratory of Induced Cellular Processes, The Federal Research Center Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences , Novosibirsk, Russia
| | - Vyacheslav Stupak
- 2 Department of Neurosurgery, Novosibirsk Research Institute of Traumatology and Orthopedics named after Ya.L. Zivian , Novosibirsk, Russia
| | - Evgeniya Voronina
- 5 Laboratory of Morphological and Molecular Biology Techniques, Regional Center of High Medical Technologies , Novosibirsk, Russia
| | - Sergey Bogachev
- 4 Laboratory of Induced Cellular Processes, The Federal Research Center Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences , Novosibirsk, Russia
| | - Ekaterina Shevela
- 1 Laboratory of Cellular Immunotherapy, Institute of Fundamental and Clinical Immunology , Novosibirsk, Russia
| | - Alexander Ostanin
- 1 Laboratory of Cellular Immunotherapy, Institute of Fundamental and Clinical Immunology , Novosibirsk, Russia
| | - Elena Chernykh
- 1 Laboratory of Cellular Immunotherapy, Institute of Fundamental and Clinical Immunology , Novosibirsk, Russia
| |
Collapse
|
20
|
Hu Z, Ma Y, Shang Z, Hu S, Liang K, Liang W, Xing X, Wang Y, Du X. Improving immunotherapy for colorectal cancer using dendritic cells combined with anti-programmed death-ligand in vitro. Oncol Lett 2018; 15:5345-5351. [PMID: 29552177 DOI: 10.3892/ol.2018.7978] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 11/29/2017] [Indexed: 12/12/2022] Open
Abstract
Monoclonal antibodies recognizing programmed death-ligand 1 (PD-L1) have been used for the clinical treatment of diverse tumor types as a form of immune checkpoint inhibitor, with a favorable therapeutic effect. Dendritic cells (DCs) are potent antigen-presenting cells that serve a pivotal role in the activation of T cells, particularly cytotoxic T lymphocytes (CTLs). DC vaccines loaded with tumor antigens, DC-CTLs and activated T cells have been revealed to be a safe and effective treatment approach against colorectal cancer within a clinical setting. In addition to tumor cells, PD-L1 is also highly expressed on DCs. As research examining the association between anti-PD-L1 and DCs is lacking, the present study compared the expression of PD-L1 on DCs in the peripheral blood of healthy donors and patients with colorectal cancer. Following the application of anti-PD-L1, the DC phenotypes, function of DC-mediated T cell induction and the cytotoxicity of CTLs were investigated by flow cytometry. The present study revealed that treatment with anti-PD-L1 may promote the maturation of DCs and enhance the functionality of the DC1 subtype. It may also increase the number of CTLs that are activated and produce CTL cells with more potent anti-tumor activity. Therefore, the creation of DC vaccines in conjunction with anti-PD-L1 may be an effective future treatment strategy for patients with colorectal cancer.
Collapse
Affiliation(s)
- Zilong Hu
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Yue Ma
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Zhiyang Shang
- Department of Tumor Prevention and Rehabilitation, PKU Care Rehabilitation Hospital, Beijing 102206, P.R. China
| | - Shidong Hu
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Kai Liang
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Wentao Liang
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Xiaowei Xing
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Yufeng Wang
- Department of Patient Admission Management, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Xiaohui Du
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| |
Collapse
|
21
|
A modified HLA-A*0201-restricted CTL epitope from human oncoprotein (hPEBP4) induces more efficient antitumor responses. Cell Mol Immunol 2018; 15:768-781. [PMID: 29375131 DOI: 10.1038/cmi.2017.155] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 11/17/2017] [Accepted: 11/17/2017] [Indexed: 12/26/2022] Open
Abstract
We previously identified human phosphatidylethanolamine-binding protein 4 (hPEBP4) as an antiapoptotic protein with increased expression levels in breast, ovarian and prostate cancer cells, but low expression levels in normal tissues, which makes hPEBP4 an attractive target for immunotherapy. Here, we developed hPEBP4-derived immunogenic peptides for inducing antigen-specific cytotoxic T lymphocytes (CTLs) targeting breast cancer. A panel of hPEBP4-derived peptides predicted by peptide-MHC-binding algorithms was evaluated to characterize their HLA-A2.1 affinity and immunogenicity. We identified a novel immunogenic peptide, P40-48 (TLFCQGLEV), that was capable of eliciting specific CTL responses in HLA-A2.1/Kb transgenic mice, as well as in peripheral blood lymphocytes from breast cancer patients. Furthermore, amino-acid substitutions in the P40-48 sequence improved its immunogenicity against hPEBP4, a self-antigen, thus circumventing tolerance. We designed peptide analogs by preferred auxiliary HLA-A*0201 anchor residue replacement, which induced CTLs that were crossreactive to the native peptide. Several analogs were able to stably bind to HLA-A*0201 and elicit specific CTL responses better than the native sequence. Importantly, adoptive transfer of CTLs induced by vaccination with two analogs more effectively inhibited tumor growth than the native peptide. These data indicate that peptide analogs with high immunogenicity represent promising candidates for peptide-mediated therapeutic cancer vaccines.
Collapse
|
22
|
Interferon-α-inducible Dendritic Cells Matured with OK-432 Exhibit TRAIL and Fas Ligand Pathway-mediated Killer Activity. Sci Rep 2017; 7:42145. [PMID: 28191816 PMCID: PMC5304184 DOI: 10.1038/srep42145] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 01/05/2017] [Indexed: 02/07/2023] Open
Abstract
Active human dendritic cells (DCs), which efficiently induce immune responses through their functions as antigen-presenting cells, exhibit direct anti-tumour killing activity in response to some pathogens and cytokines. These antigen-presenting and tumour killing abilities may provide a breakthrough in cancer immunotherapy. However, the mechanisms underlying this killer DC activity have not been fully proven, despite the establishment of interferon-α (IFN-α)-generated killer DCs (IFN-DCs). Here mature IFN-DCs (mIFN-DCs), generated from IFN-DCs primed with OK-432 (streptococcal preparation), exhibited elevated expression of CD86 and human leukocyte antigen-DR (minimum criteria for DC vaccine clinical trials) as well as antigen-presenting abilities comparable with those of mature IL-4-DCs (mIL-4-DCs). Interestingly, the killing activity of mIFN-DCs, which correlated with the expression of CD56 (natural killer cell marker) and was activated via the tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) and Fas ligand pathway, was stronger than that of IFN-DCs and remarkably stronger than that of mIL-4-DCs. Therefore, mIFN-DCs exhibit great potential as an anti-cancer vaccine that would promote both acquired immunity and direct tumour killing.
Collapse
|
23
|
Dendritic cells in hematological malignancies. Crit Rev Oncol Hematol 2016; 108:86-96. [DOI: 10.1016/j.critrevonc.2016.10.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 09/05/2016] [Accepted: 10/18/2016] [Indexed: 01/17/2023] Open
|
24
|
Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand-Induced Apoptosis in Prostate Cancer Cells after Treatment with Xanthohumol-A Natural Compound Present in Humulus lupulus L. Int J Mol Sci 2016; 17:ijms17060837. [PMID: 27338375 PMCID: PMC4926371 DOI: 10.3390/ijms17060837] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 05/19/2016] [Accepted: 05/23/2016] [Indexed: 01/31/2023] Open
Abstract
TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) is an endogenous ligand, which plays role in immune surveillance and anti-tumor immunity. It has ability to selectively kill tumor cells showing no toxicity to normal cells. We tested the apoptotic and cytotoxic activities of xanthohumol, a prenylated chalcone found in Humulus lupulus on androgen-sensitive human prostate adenocarcinoma cells (LNCaP) in combination with TRAIL. Cytotoxicity was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide tetrazolium reduction assay (MTT) and lactate dehydrogenase assay (LDH). The expression of death receptors (DR4/TRAIL-R1 and DR5/TRAIL-R2) and apoptosis were detected using flow cytometry. We examined mitochondrial membrane potential (ΔΨm) by DePsipher reagent using fluorescence microscopy. The intracellular expression of proteins was evaluated by Western blotting. Our study showed that xanthohumol enhanced cytotoxic and apoptotic effects of TRAIL. The tested compounds activated caspases-3, -8, -9, Bid, and increased the expression of Bax. They also decreased expression of Bcl-xL and decreased mitochondrial membrane potential, while the expression of death receptors was not changed. The findings suggest that xanthohumol is a compound of potential use in chemoprevention of prostate cancer due to its sensitization of cancer cells to TRAIL-mediated apoptosis.
Collapse
|
25
|
Li X, Liu X, Zhao Y, Zhong R, Song A, Sun L. Effect of thymosin α₁ on the phenotypic and functional maturation of dendritic cells from children with acute lymphoblastic leukemia. Mol Med Rep 2015; 12:6093-7. [PMID: 26239360 DOI: 10.3892/mmr.2015.4153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 06/15/2015] [Indexed: 11/06/2022] Open
Abstract
To determine the effect of thymosin α1 (Tα1) on the phenotypic and functional maturation of HL‑60 cells, freeze‑thaw antigen‑loaded dendritic cells (DCs) were derived from peripheral blood mononuclear cells (PBMCs) of children with acute lymphoblastic leukemia (ALL). The DCs were generated from the PBMC samples that were collected from the PB of 10 consecutive ALL children. On day 3 of culturing, the cells in the antigen + no Tα1 (AN) and antigen + Tα1 (AT) groups were incubated with 100 µl lysates obtained from freeze‑thaw cycling. After 5 days of incubation, the AT group was administered with 100 ng/ml Tα1. On day 8, the DCs were stained with fluorescein isothiocyanate‑conjugated cluster of differentiation (CD)1a, CD83 and HLA‑DR antibodies and analyzed by flow cytometry. In addition, the killing activity of cytotoxic T lymphocytes (CTLs) from the different groups on wild‑type leukemia cells was measured. The DCs in the AT group exhibited more apparent, characteristic dendritic morphologies than the control and AN group DCs. Furthermore, the lowest expression level of CD1a, and the highest expression of CD83 and HLA‑DR were observed in the AT group when compared with the AN and control groups (P<0.05). The lactate dehydrogenase release assay demonstrated that the killing rate of CTL in the AT group was significantly higher than that in the control and AN groups (P<0.01). Thus, Tα1 may markedly promote the phenotypic and functional maturation of DCs, and may serve as a suitable immunomodulator of DC‑based immunotherapy for treatment of hematological malignancies.
Collapse
Affiliation(s)
- Xuerong Li
- Department of Pediatric Hematology and Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Xiaodan Liu
- Department of Pediatric Hematology and Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Yanxia Zhao
- Department of Pediatric Hematology and Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Ren Zhong
- Department of Pediatric Hematology and Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Aiqin Song
- Department of Pediatric Hematology and Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Lirong Sun
- Department of Pediatric Hematology and Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
26
|
Sun W, Wei X, Niu A, Ma X, Li JJ, Gao D. Enhanced anti-colon cancer immune responses with modified eEF2-derived peptides. Cancer Lett 2015; 369:112-23. [PMID: 26304717 DOI: 10.1016/j.canlet.2015.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/31/2015] [Accepted: 08/05/2015] [Indexed: 12/22/2022]
Abstract
Eukaryotic elongation factor-2 (eEF2) is overexpressed in many human cancers and is an attractive target for cancer immunotherapy. The eEF2 derived polypeptides have been shown to be able to induce cytotoxic T lymphocytes from healthy donor. Here, we demonstrate the evidence indicating that modification of a segment of peptides from wild type eEF2-derived immunogenic peptides is able to further enhance its capacity of inducing antigen-specific cytotoxic T lymphocytes (CTLs) against colon cancer cells. Using peptide-MHC binding algorithms, potential HLA-A2.1-restricted epitopes capable of inducing specific CD8(+) CTLs were identified. By analyzing HLA-A2.1 affinity and immunogenicity, we further identified one novel immunogenic peptide, P739-747 (RLMEPIYLV), that elicited specific CTL responses in HLA-A2.1/K(b) transgenic mice and culture with peripheral blood lymphocytes from colon cancer patients. Furthermore, replacing certain amino acids (at positions 1, 3, 7) within the P739-747 sequence improved the immunogenicity against eEF2. Several analogs containing the auxiliary HLA-A*0201 anchor residues were able to stably bind to HLA-A*0201 and enhance CTL responses compared with the native sequence; two of them showed increased anti-tumor effects during the adoptive immunotherapy in vivo. Thus, these results support that modified immunogenic analogs are promising candidates for peptide-based cancer vaccination and immunotherapy.
Collapse
Affiliation(s)
- Weihong Sun
- Biotherapy Center, The Second Affiliated Hospital, Qingdao University Medical College, Qingdao 266042, China.
| | - Xiaofang Wei
- Biotherapy Center, The Second Affiliated Hospital, Qingdao University Medical College, Qingdao 266042, China
| | - Airong Niu
- Department of Clinical Laboratory, The Second Affiliated Hospital, Qingdao University Medical College, Qingdao 266042, China
| | - Xuezhen Ma
- Department of Oncology, The Second Affiliated Hospital, Qingdao University Medical College, Qingdao 266042, China
| | - Jian Jian Li
- Department of Radiation Oncology, NCI-designated Compressive Cancer Center, University of California Davis, Sacramento, CA 95817, USA.
| | - Daiqing Gao
- Biotherapy Center, The Second Affiliated Hospital, Qingdao University Medical College, Qingdao 266042, China
| |
Collapse
|
27
|
Hira SK, Mondal I, Bhattacharya D, Gupta KK, Manna PP. Downregulation of STAT3 phosphorylation enhances tumoricidal effect of IL-15-activated dendritic cell against doxorubicin-resistant lymphoma and leukemia via TNF-α. Int J Biochem Cell Biol 2015; 67:1-13. [PMID: 26255115 DOI: 10.1016/j.biocel.2015.08.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 05/30/2015] [Accepted: 08/03/2015] [Indexed: 12/11/2022]
Abstract
Although disputed by some, increasing evidence suggests that TNF-α synergies with traditional chemotherapeutic drugs to exert a heightened antitumor effect. The present study investigated the antitumor efficacy of recombinant IL-15 in combination with the STAT3 inhibitor cucurbitacin-I in a doxorubicin-resistant murine lymphoma model. The significance of the work is to understand and design effective strategies in doxorubicin resistant lymphomas. TNF-α is downregulated in dendritic cells from mice with Dalton's lymphoma and shows an inverse relationship with disease progression. Doxorubicin-resistant DL cells have elevated levels of Bcl-2 and Mcl-1 and increased phosphorylation of STAT3. These cells are refractory to dendritic cell derived TNF-α. Doxorubicin resistant Dalton's lymphoma is susceptible to dendritic cell derived TNF-α upon stimulation with the STAT3 inhibitor cucurbitacin-I, which downregulates STAT3 and other survival molecules. The combined treatment of low dose of cucurbitacin-I and rIL-15 is ineffective in mice with doxorubicin resistant Dalton's lymphoma, but a similar therapy prolongs the survival of mice transplanted with parental Dalton's lymphoma. Doxorubicin resistant Dalton's lymphoma responds to therapy with high doses of cucurbitacin-I and rIL-15. Dendritic cell derived from mice responded positively to the therapy and regained their tumoricidal properties with respect to growth inhibition and killing of DL tumor cells. Similar to DL, DC derived from CML patients are impaired in TNF-α expression and are unable to restrict the growth of drug-resistant lymphoma and leukemia cells. This combination approach could be used as a new therapeutic strategy for aggressive and highly metastatic doxorubicin resistant lymphoma.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Coculture Techniques
- Cytotoxicity, Immunologic
- Dendritic Cells/drug effects
- Dendritic Cells/metabolism
- Dendritic Cells/pathology
- Disease Models, Animal
- Doxorubicin/pharmacology
- Drug Resistance, Neoplasm
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Interleukin-15/pharmacology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Lymphocytes/drug effects
- Lymphocytes/metabolism
- Lymphocytes/pathology
- Lymphoma/drug therapy
- Lymphoma/metabolism
- Lymphoma/mortality
- Lymphoma/pathology
- Mice
- Mice, Inbred AKR
- Myeloid Cell Leukemia Sequence 1 Protein/genetics
- Myeloid Cell Leukemia Sequence 1 Protein/metabolism
- Phosphorylation
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Recombinant Proteins/pharmacology
- STAT3 Transcription Factor/antagonists & inhibitors
- STAT3 Transcription Factor/genetics
- STAT3 Transcription Factor/metabolism
- Signal Transduction
- Survival Analysis
- Triterpenes/pharmacology
- Tumor Necrosis Factor-alpha/agonists
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Sumit Kumar Hira
- Immunobiology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, Uttar Pradesh, India; Department of Zoology, The University of Burdwan, Burdwan, West Bengal, India
| | - Indrani Mondal
- Department of Pathology, Burdwan Medical College, Burdwan, West Bengal, India; Department of Pathology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | | | - Kailash Kumar Gupta
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Partha Pratim Manna
- Immunobiology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, Uttar Pradesh, India.
| |
Collapse
|
28
|
Pandey VK, Amin PJ, Shankar BS. G1-4A, a polysaccharide from Tinospora cordifolia induces peroxynitrite dependent killer dendritic cell (KDC) activity against tumor cells. Int Immunopharmacol 2014; 23:480-8. [DOI: 10.1016/j.intimp.2014.09.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 09/15/2014] [Accepted: 09/17/2014] [Indexed: 11/30/2022]
|
29
|
Leplina OY, Tyrinova TV, Tikhonova MA, Ostanin AA, Chernykh ER. Interferon alpha induces generation of semi-mature dendritic cells with high pro-inflammatory and cytotoxic potential. Cytokine 2014; 71:1-7. [PMID: 25174880 DOI: 10.1016/j.cyto.2014.07.258] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 07/31/2014] [Indexed: 01/04/2023]
Abstract
Dendritic cell-based vaccines are considered as a new and promising immunotherapeutic approach for cancer treatment. However, the choice of optimal protocol of dendritic cell generation in vitro represents the major challenge. Here, we compared phenotype and functional characteristics of human monocyte-derived dendritic cells (DCs) generated in the presence of IL-4/GM-CSF (IL4-DCs) and IFNα/GM-CSF (IFN-DCs). We showed that IFN-DCs displayed semi-mature phenotype and expressed higher level of CD123, TNF-related apoptosis-inducing ligand (TRAIL) and B7-H1 molecules in comparison with IL4-DCs. LPS-stimulated IFN-DCs were characterized by greater production of Th1/pro-inflammatory (IFN-γ, IL-2, IL-1β, TNF-α, IL-17), Тh2/anti-inflammatory cytokines (IL-10, IL-5), hematopoietic growth factors (G-CSF) and chemokines (MCP-1). These data indicated more pronounced ability of IFN-DCs to induce cellular immune response as well as humoral immune response compared to IL4-DCs. LPS-stimulated IFN-DCs possessed higher direct cytotoxic activity against TRAIL-sensitive tumor cell line Jurkat and similar cytotoxicity against TRAIL-resistant tumor HEp-2 cells. Besides, IFN-DCs and IL4-DCs equally induced apoptosis of activated CD4(+) and CD8(+) T cells. These results suggest that IFN-DCs can be used as potent cell-based curative therapies for individuals with cancer.
Collapse
Affiliation(s)
- Olga Yu Leplina
- Institute of Clinical Immunology SB RAMS, 14 Yadrintsevskaya Str., 630099 Novosibirsk, Russia(1)
| | - Tamara V Tyrinova
- Institute of Clinical Immunology SB RAMS, 14 Yadrintsevskaya Str., 630099 Novosibirsk, Russia(1).
| | - Marina A Tikhonova
- Institute of Clinical Immunology SB RAMS, 14 Yadrintsevskaya Str., 630099 Novosibirsk, Russia(1)
| | - Alexander A Ostanin
- Institute of Clinical Immunology SB RAMS, 14 Yadrintsevskaya Str., 630099 Novosibirsk, Russia(1)
| | - Elena R Chernykh
- Institute of Clinical Immunology SB RAMS, 14 Yadrintsevskaya Str., 630099 Novosibirsk, Russia(1)
| |
Collapse
|
30
|
Hanke NT, LaCasse CJ, Larmonier CB, Alizadeh D, Trad M, Janikashvili N, Bonnotte B, Katsanis E, Larmonier N. PIAS1 and STAT-3 impair the tumoricidal potential of IFN-γ-stimulated mouse dendritic cells generated with IL-15. Eur J Immunol 2014; 44:2489-2499. [PMID: 24777831 DOI: 10.1002/eji.201343803] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 04/04/2014] [Accepted: 04/17/2014] [Indexed: 01/22/2023]
Abstract
Primarily defined by their antigen-presenting property, dendritic cells (DCs) are being implemented as cancer vaccines in immunotherapeutic interventions. DCs can also function as direct tumor cell killers. How DC cytotoxic activity can be efficiently harnessed and the mechanisms controlling this nonconventional property are not fully understood. We report here that the tumoricidal potential of mouse DCs generated from myeloid precursors with GM-CSF and IL-15 (IL-15 DCs) can be triggered with the Toll-like receptor (TLR) 4 ligand lipopolysaccharide to a similar extent compared with that of their counterparts, conventionally generated with IL-4 (IL-4 DCs). The mechanism of tumor cell killing depends on the induction of iNOS expression by DCs. In contrast, interferon (IFN)-γ induces the cytotoxic activity of IL-4 but not IL-15 DCs. Although the IFN-γ-STAT-1 signaling pathway is overall functional in IL-15 DCs, IFN-γ fails to induce iNOS expression in these cells. iNOS expression is negatively controlled in IFN-γ-stimulated IL-15 DCs by the cooperation between the E3 SUMO ligase PIAS1 and STAT-3, and can be partially restored with PIAS1 siRNA and STAT-3 inhibitors.
Collapse
Affiliation(s)
- Neale T Hanke
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States of America.,Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, United States of America
| | - Collin J LaCasse
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States of America.,Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, United States of America
| | - Claire B Larmonier
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States of America
| | - Darya Alizadeh
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States of America.,Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, United States of America
| | - Malika Trad
- INSERM UMR 1098, Faculty of Medicine, Dijon, France
| | | | | | - Emmanuel Katsanis
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States of America.,Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, United States of America.,Department of Immunobiology, BIO5 Institute and Arizona Cancer Center, University of Arizona, Tucson, AZ, United States of America
| | - Nicolas Larmonier
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States of America.,Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, United States of America.,Department of Immunobiology, BIO5 Institute and Arizona Cancer Center, University of Arizona, Tucson, AZ, United States of America
| |
Collapse
|
31
|
Wang P, Xue Y, Han Y, Lin L, Wu C, Xu S, Jiang Z, Xu J, Liu Q, Cao X. The STAT3-binding long noncoding RNA lnc-DC controls human dendritic cell differentiation. Science 2014; 344:310-3. [PMID: 24744378 DOI: 10.1126/science.1251456] [Citation(s) in RCA: 858] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Long noncoding RNAs (lncRNAs) play important roles in diverse biological processes; however, few have been identified that regulate immune cell differentiation and function. Here, we identified lnc-DC, which was exclusively expressed in human conventional dendritic cells (DCs). Knockdown of lnc-DC impaired DC differentiation from human monocytes in vitro and from mouse bone marrow cells in vivo and reduced capacity of DCs to stimulate T cell activation. lnc-DC mediated these effects by activating the transcription factor STAT3 (signal transducer and activator of transcription 3). lnc-DC bound directly to STAT3 in the cytoplasm, which promoted STAT3 phosphorylation on tyrosine-705 by preventing STAT3 binding to and dephosphorylation by SHP1. Our work identifies a lncRNA that regulates DC differentiation and also broadens the known mechanisms of lncRNA action.
Collapse
Affiliation(s)
- Pin Wang
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Tel J, Anguille S, Waterborg CEJ, Smits EL, Figdor CG, de Vries IJM. Tumoricidal activity of human dendritic cells. Trends Immunol 2013; 35:38-46. [PMID: 24262387 PMCID: PMC7106406 DOI: 10.1016/j.it.2013.10.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 10/18/2013] [Accepted: 10/21/2013] [Indexed: 12/11/2022]
Abstract
Human DC subsets can exert tumoricidal activity. Killer DCs exploit several mechanisms for direct killing of target cells, including TRAIL and granzyme B. Antigen presentation and/or IFN production are important additional effector functions. Killer DCs are promising targets for immunotherapeutic strategies.
Dendritic cells (DCs) are a family of professional antigen-presenting cells (APCs) that are able to initiate innate and adaptive immune responses against pathogens and tumor cells. The DC family is heterogeneous and is classically divided into two main subsets, each with its unique phenotypic and functional characteristics: myeloid DCs (mDCs) and plasmacytoid DCs (pDCs). Recent results have provided intriguing evidence that both DC subsets can also function as direct cytotoxic effector cells; in particular, against cancer cells. In this review, we delve into this understudied function of human DCs and discuss why these so-called killer DCs might become important tools in future cancer immunotherapies.
Collapse
Affiliation(s)
- Jurjen Tel
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Sébastien Anguille
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Claire E J Waterborg
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Evelien L Smits
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium; Center for Oncological Research, University of Antwerp, Antwerp, Belgium
| | - Carl G Figdor
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands; Department of Medical Oncology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| |
Collapse
|
33
|
Diao Z, Shi J, Zhu J, Yuan H, Ru Q, Liu S, Liu Y, Zheng D. TRAIL suppresses tumor growth in mice by inducing tumor-infiltrating CD4(+)CD25 (+) Treg apoptosis. Cancer Immunol Immunother 2013; 62:653-63. [PMID: 23143747 PMCID: PMC11028869 DOI: 10.1007/s00262-012-1370-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 10/31/2012] [Indexed: 01/22/2023]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), a promising and novel anticancer cytokine, specifically kills numerous tumor cells by apoptosis. However, some malignancies are resistant to TRAIL treatment in clinical trials, thus limiting its therapeutic potential. In the present study, the TRAIL-resistant murine hepatocellular carcinoma cell line Hepa1-6 was used to elucidate the physiological significance of TRAIL resistance, especially with respect to the immune regulatory function of TRAIL. Hepa1-6 cells were resistant to TRAIL-induced apoptosis in vitro; however, intratumoral injection of recombinant soluble TRAIL inhibited tumor growth and prolonged survival time in tumor-bearing mice. Local TRAIL treatment decreased the number of intratumoral CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs) but did not affect CD4(+)CD25(+)Foxp3(+) Tregs in the draining lymph nodes and spleen. Further investigation showed that TRAIL induced apoptosis of tumor-activated CD4(+)CD25(+)Foxp3(+) Tregs, but not of CD4(+)CD25(-) T cells. Moreover, mouse TRAIL receptor DR5 expression was detected on the surface of the tumor-infiltrating CD4(+)CD25(+)Foxp3(+) Tregs, but not on naïve CD4(+)CD25(+)Foxp3(+) Tregs. Interestingly, intratumoral injection of TRAIL not only decreased the number of CD4(+)CD25(+)Foxp3(+) Tregs but also increased the number of tumor-specific CD8(+) CTL and augmented their cytotoxicity to the tumor cells. These data provide the novel evidence for an immune regulatory function of TRAIL and may shed light on the clinical application of TRAIL.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Cell Growth Processes/drug effects
- Cell Line, Tumor
- Female
- Interleukin-2 Receptor alpha Subunit/immunology
- Liver Neoplasms, Experimental/drug therapy
- Liver Neoplasms, Experimental/immunology
- Liver Neoplasms, Experimental/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- TNF-Related Apoptosis-Inducing Ligand/immunology
- TNF-Related Apoptosis-Inducing Ligand/pharmacology
Collapse
Affiliation(s)
- Zhijuan Diao
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005 China
| | - Juan Shi
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005 China
| | - Jieqing Zhu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005 China
| | - Haiqin Yuan
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005 China
| | - Qiang Ru
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005 China
| | - Shilian Liu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005 China
| | - Yanxin Liu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005 China
| | - Dexian Zheng
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing, 100005 China
| |
Collapse
|
34
|
Högner K, Wolff T, Pleschka S, Plog S, Gruber AD, Kalinke U, Walmrath HD, Bodner J, Gattenlöhner S, Lewe-Schlosser P, Matrosovich M, Seeger W, Lohmeyer J, Herold S. Macrophage-expressed IFN-β contributes to apoptotic alveolar epithelial cell injury in severe influenza virus pneumonia. PLoS Pathog 2013; 9:e1003188. [PMID: 23468627 PMCID: PMC3585175 DOI: 10.1371/journal.ppat.1003188] [Citation(s) in RCA: 177] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 12/27/2012] [Indexed: 12/31/2022] Open
Abstract
Influenza viruses (IV) cause pneumonia in humans with progression to lung failure and fatal outcome. Dysregulated release of cytokines including type I interferons (IFNs) has been attributed a crucial role in immune-mediated pulmonary injury during severe IV infection. Using ex vivo and in vivo IV infection models, we demonstrate that alveolar macrophage (AM)-expressed IFN-β significantly contributes to IV-induced alveolar epithelial cell (AEC) injury by autocrine induction of the pro-apoptotic factor TNF-related apoptosis-inducing ligand (TRAIL). Of note, TRAIL was highly upregulated in and released from AM of patients with pandemic H1N1 IV-induced acute lung injury. Elucidating the cell-specific underlying signalling pathways revealed that IV infection induced IFN-β release in AM in a protein kinase R- (PKR-) and NF-κB-dependent way. Bone marrow chimeric mice lacking these signalling mediators in resident and lung-recruited AM and mice subjected to alveolar neutralization of IFN-β and TRAIL displayed reduced alveolar epithelial cell apoptosis and attenuated lung injury during severe IV pneumonia. Together, we demonstrate that macrophage-released type I IFNs, apart from their well-known anti-viral properties, contribute to IV-induced AEC damage and lung injury by autocrine induction of the pro-apoptotic factor TRAIL. Our data suggest that therapeutic targeting of the macrophage IFN-β-TRAIL axis might represent a promising strategy to attenuate IV-induced acute lung injury.
Collapse
MESH Headings
- Acute Lung Injury/immunology
- Acute Lung Injury/metabolism
- Acute Lung Injury/pathology
- Adult
- Animals
- Apoptosis
- Disease Models, Animal
- Humans
- Influenza, Human/immunology
- Influenza, Human/metabolism
- Influenza, Human/pathology
- Interferon-beta/metabolism
- Macrophages, Alveolar/immunology
- Macrophages, Alveolar/metabolism
- Macrophages, Alveolar/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Mosaicism
- Pneumonia, Viral/immunology
- Pneumonia, Viral/metabolism
- Pneumonia, Viral/pathology
- Respiratory Mucosa/immunology
- Respiratory Mucosa/metabolism
- Respiratory Mucosa/pathology
- TNF-Related Apoptosis-Inducing Ligand/metabolism
Collapse
Affiliation(s)
- Katrin Högner
- Department of Internal Medicine II, University of Giessen Lung Center (UGLC) and German Center for Lung Research (DZL), Giessen, Germany
| | - Thorsten Wolff
- Division of Influenza/Respiratory Viruses, Robert Koch-Institute, Berlin, Germany
| | - Stephan Pleschka
- Institute of Medical Virology, Justus-Liebig-University, Giessen, Germany
| | - Stephanie Plog
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Achim D. Gruber
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, Twincore Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Hans-Dieter Walmrath
- Department of Internal Medicine II, University of Giessen Lung Center (UGLC) and German Center for Lung Research (DZL), Giessen, Germany
| | - Johannes Bodner
- Division of Thoracic Surgery, University of Giessen Lung Center (UGLC) and German Center for Lung Research (DZL), Giessen, Germany
| | - Stefan Gattenlöhner
- Department of Pathology, University of Giessen Lung Center (UGLC) and German Center for Lung Research (DZL), Giessen, Germany
| | - Peter Lewe-Schlosser
- Center for Radiation Therapy, University of Giessen Lung Center (UGLC) and German Center for Lung Research (DZL), Giessen, Germany
| | | | - Werner Seeger
- Department of Internal Medicine II, University of Giessen Lung Center (UGLC) and German Center for Lung Research (DZL), Giessen, Germany
| | - Juergen Lohmeyer
- Department of Internal Medicine II, University of Giessen Lung Center (UGLC) and German Center for Lung Research (DZL), Giessen, Germany
| | - Susanne Herold
- Department of Internal Medicine II, University of Giessen Lung Center (UGLC) and German Center for Lung Research (DZL), Giessen, Germany
- * E-mail:
| |
Collapse
|
35
|
Neve A, Corrado A, Cantatore FP. TNF-related apoptosis-inducing ligand (TRAIL) in rheumatoid arthritis: what's new? Clin Exp Med 2012; 14:115-20. [PMID: 23275079 DOI: 10.1007/s10238-012-0226-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 12/20/2012] [Indexed: 01/19/2023]
Abstract
TNF-related apoptosis-inducing ligand (TRAIL) is a type II transmembrane protein of the TNF superfamily that serves as an extracellular signal that triggers programmed cell death in tumor cells, without affecting normal cells. Recently, scientists have turned their attention to the emerging role of TRAIL in immune and autoimmune responses. TRAIL has been shown to down-regulate the self-antigens in autoimmune diseases, such as rheumatoid arthritis (RA) by exerting its apoptotic effect on activated T cells and synoviocytes and by its local anti-inflammatory effect. The impact of TRAIL molecular variants and agonistic monoclonal antibodies in the regulation of TRAIL activity in arthritis animal models strongly supports the idea of testing the role of TRAIL in humans, with the aim of developing new effective therapies that promote apoptosis of synoviocytes and/or infiltrating lymphocytes, by targeting TRAIL. The aim of this review is to summarize recent progress and current knowledge of TRAIL functions in RA.
Collapse
Affiliation(s)
- Anna Neve
- Department of Medical and Surgical Sciences, Rheumatology Clinic, University of Foggia, Ospedale "Col. D'Avanzo", V.le degli Aviatori 1, 71100, Foggia, Italy
| | | | | |
Collapse
|
36
|
Cytotoxic activity of dendritic cells as a possible mechanism of negative regulation of T lymphocytes in pulmonary tuberculosis. Clin Dev Immunol 2012; 2012:628635. [PMID: 23056139 PMCID: PMC3465906 DOI: 10.1155/2012/628635] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 08/23/2012] [Accepted: 08/31/2012] [Indexed: 12/16/2022]
Abstract
The PD-1/B7-H1-mediated induction of T cell apoptosis/anergy as a possible mechanism of immune response failure was studied in 76 patients with pulmonary tuberculosis (TB) with normal and low-proliferative response to antigens of M. tuberculosis (purified protein derivative (PPD)). It was revealed that dendritic cells (DCs), generated in vitro from patient blood monocytes with GM-CSF + IFN-α, were characterized by increased B7-H1 expression, upproduction of IL-10, and reducing of allostimulatory activity in mixed lymphocyte culture (MLC). Moreover, DCs of patients with TB were able to enhance T cell apoptosis and to block T-cell division in MLC. It was shown that neutralizing anti-PD1 antibodies significantly decreased the proapoptogenic/tolerogenic effect of DCs. Correlation analysis revealed a direct relationship between IL-10 production and level of B7-H1 expression in the general group of investigated patients. It was demonstrated that generation of healthy donor DCs in the presence of IL-10 led to an increase in the number of DCs-expressed B7-H1 molecule, DC proapoptogenic activity, and a decrease in their allostimulatory activity. Obviously, the revealed phenomenon of the PD-1/B7-H1-mediated pro-apoptogenic activity of DCs is clinically significant since the cytotoxic/tolerogenic potential of DCs is more pronounced in patients with PPD anergy.
Collapse
|
37
|
LaCasse CJ, Janikashvili N, Larmonier CB, Alizadeh D, Hanke N, Kartchner J, Situ E, Centuori S, Har-Noy M, Bonnotte B, Katsanis E, Larmonier N. Th-1 lymphocytes induce dendritic cell tumor killing activity by an IFN-γ-dependent mechanism. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 187:6310-7. [PMID: 22075702 PMCID: PMC3297475 DOI: 10.4049/jimmunol.1101812] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Dendritic cells (DCs) encompass a heterogeneous population of cells capable of orchestrating innate and adaptive immune responses. The ability of DCs to act as professional APCs has been the foundation for the development and use of these cells as vaccines in cancer immunotherapy. DCs are also endowed with the nonconventional property of directly killing tumor cells. The current study investigates the regulation of murine DC cytotoxic function by T lymphocytes. We provide evidence that CD4(+) Th-1, but not Th-2, Th-17 cells, or regulatory T cells, are capable of inducing DC cytotoxic function. IFN-γ was identified as the major factor responsible for Th-1-induced DC tumoricidal activity. Tumor cell killing mediated by Th-1-activated killer DCs was dependent on inducible NO synthase expression and NO production. Importantly, Th-1-activated killer DCs were capable of presenting the acquired Ags from the killed tumor cells to T lymphocytes in vitro or in vivo. These observations offer new possibilities for the application of killer DCs in cancer immunotherapy.
Collapse
MESH Headings
- Animals
- Antigen Presentation/genetics
- Antigen Presentation/immunology
- Bone Marrow Cells/cytology
- Bone Marrow Cells/immunology
- Bone Marrow Cells/metabolism
- Cell Line, Tumor
- Coculture Techniques
- Cytotoxicity, Immunologic/genetics
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Female
- Interferon-gamma/metabolism
- Interferon-gamma/physiology
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/therapy
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice
- Mice, 129 Strain
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Receptors, Interferon/deficiency
- Receptors, Interferon/genetics
- Receptors, Interferon/physiology
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Interferon gamma Receptor
Collapse
Affiliation(s)
- Collin J. LaCasse
- Department of Pediatrics, University of Arizona, Tucson, AZ 85724-5073
- Department of Immunobiology, University of Arizona, Tucson, AZ 85724-5073
| | - Nona Janikashvili
- Department of Pediatrics, University of Arizona, Tucson, AZ 85724-5073
| | | | - Darya Alizadeh
- Department of Pediatrics, University of Arizona, Tucson, AZ 85724-5073
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85724-5073
| | - Neale Hanke
- Department of Pediatrics, University of Arizona, Tucson, AZ 85724-5073
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85724-5073
| | - Jessica Kartchner
- Department of Pediatrics, University of Arizona, Tucson, AZ 85724-5073
| | - Elaine Situ
- Department of Pediatrics, University of Arizona, Tucson, AZ 85724-5073
| | - Sara Centuori
- Department of Pediatrics, University of Arizona, Tucson, AZ 85724-5073
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85724-5073
| | - Michael Har-Noy
- Immunovative Therapies Ltd, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | - Emmanuel Katsanis
- Department of Pediatrics, University of Arizona, Tucson, AZ 85724-5073
- Department of Immunobiology, University of Arizona, Tucson, AZ 85724-5073
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85724-5073
- BIO5 Institute and Arizona Cancer Center, University of Arizona, Tucson, AZ 85724-5073
| | - Nicolas Larmonier
- Department of Pediatrics, University of Arizona, Tucson, AZ 85724-5073
- Department of Immunobiology, University of Arizona, Tucson, AZ 85724-5073
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85724-5073
- BIO5 Institute and Arizona Cancer Center, University of Arizona, Tucson, AZ 85724-5073
| |
Collapse
|
38
|
Iyer N, Marathe SA, Chaudhuri D, Garai P, Chakravortty D. Immunomodulation using agonists and antagonists: potential clinical applications. Expert Opin Investig Drugs 2011; 21:67-81. [PMID: 22149716 DOI: 10.1517/13543784.2012.642367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Extensive studies have gone into understanding the differential role of the innate and adaptive arms of the immune system in the context of various diseases. Receptor-ligand interactions are responsible for mediating cross-talk between the innate and adaptive arms of the immune system, so as to effectively counter the pathogenic challenge. While TLRs remain the best studied innate immune receptor, many other receptor families are now coming to the fore for their role in various pathologies. Research has focused on the discovery of novel agonists and antagonists for these receptors as potential therapeutics. AREAS COVERED In this review, we present an overview of the recent advances in the discovery of drugs targeting important receptors such as G-protein coupled receptors, TRAIL-R, IL-1β receptor, PPARs, etc. All these receptors play a critical role in the modulation of the immune response. We focus on the recent paradigms applied for the generation of specific and effective therapeutics for these receptors and their status in clinical trials. EXPERT OPINION Non-specific activation by antagonist/agonist is a difficult problem to dodge. This demands innovation in ligand designing with the use of strategies such as allosterism and dual-specific ligands. Rigorous preclinical and clinical studies are required in transforming a compound to a therapeutic.
Collapse
Affiliation(s)
- Namrata Iyer
- Indian Institute of Science, Centre for Infectious Disease Research and Biosafety Laboratories, Department of Microbiology and Cell Biology, Bangalore 560012, India
| | | | | | | | | |
Collapse
|
39
|
Zhong H, Yang Y, Ma S, Xiu F, Cai Z, Zhao H, Du L. Induction of a tumour-specific CTL response by exosomes isolated from heat-treated malignant ascites of gastric cancer patients. Int J Hyperthermia 2011; 27:604-11. [PMID: 21846196 DOI: 10.3109/02656736.2011.564598] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Tumour cell-derived exosomes may represent a novel type of cancer vaccine. However, the immunogenicity of exosomes derived from tumour cells has been shown to be poor. Therefore, in this study, exosome immunogenicity following heat treatment of exosomes from malignant ascites obtained from gastric cancer patients was evaluated. MATERIALS AND METHODS Tumour-derived exosomes were isolated from heat-treated and untreated malignant ascites of gastric cancer patients using serial centrifugation and sucrose gradient ultracentrifugation. Next, in vitro experiments were performed to investigate the influence of heat treatment on exosome immunogenicity. RESULTS Exosomes from heat-treated malignant ascites of gastric cancer patients (HS exosomes) were found to contain higher concentrations of heat shock proteins, Hsp70 and Hsp60, than exosomes derived from untreated malignant ascites obtained from gastric cancer patients. Additional in vitro studies suggest that exosomes derived from heat-treated malignant ascites are able to promote dendritic cell (DC) maturation and induce a tumour-specific cytotoxic T lymphocyte (CTL) response. CONCLUSIONS Overall, these results demonstrate that exposure to heat stress can improve the immunogenicity of exosomes obtained from malignant ascites of gastric cancer patients.
Collapse
Affiliation(s)
- Haijun Zhong
- Department of Chemotherapy, Zhejiang Cancer Hospital, Hangzhou, China
| | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Natural killer (NK) cells and dendritic cells (DCs) are essential effector cells of the innate immune system that rapidly recognize and eliminate microbial pathogens and abnormal cells, and induce and regulate adaptive immune functions. While NK cells express perforin and granzymes in the lysosomal granules and transmembrane tumor necrosis factor superfamily ligands (tmTNFSFL) on the plasma membrane, DCs express only tmTNFSFL on the plasma membrane. Perforin and granzymes are cytolytic molecules, which NK cells use to mediate a secretory/necrotic killing mechanism against rare leukemia cell targets. TNFSFL are pleiotropic transmembrane molecules, which can mediate a variety of important functions such as apoptosis, development of peripheral lymphoid tissues, inflammation and regulation of immune functions. Using tmTNFSFL, NK cells and DCs mediate a cell contact-dependent non-secretory apoptotic cytotoxic mechanism against virtually all types of cancer cells, and cross talk that leads to polarization and reciprocal stimulation and amplification of Th1 type cytokines secreted by NK cells and DCs. In this paper, we review and discuss the supporting evidence of the non-secretory, tmTNFSFL-mediated innate mechanisms of NK cells and DCs, their roles in anticancer immune defense and potential of their modulation and use in prevention and treatment of cancer.
Collapse
|
41
|
Kaminskyy VO, Surova OV, Vaculova A, Zhivotovsky B. Combined inhibition of DNA methyltransferase and histone deacetylase restores caspase-8 expression and sensitizes SCLC cells to TRAIL. Carcinogenesis 2011; 32:1450-8. [PMID: 21771726 DOI: 10.1093/carcin/bgr135] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising drug for the treatment of tumors; however, a number of cancer cells are resistant to this cytokine. Among the mechanisms of resistance of small cell lung carcinomas (SCLCs) to TRAIL is the lack of caspase-8 expression. Although methylation of the caspase-8 promoter has been suggested as the main mechanism of caspase-8 silencing, we showed that reduction of the enzymes involved in DNA methylation, DNA methyltransferases (DNMT) 1, 3a and 3b, was not sufficient to significantly restore caspase-8 expression in SCLC cells, signifying that other mechanisms are involved in caspase-8 silencing. We found that combination of the DNMT inhibitor decitabine with an inhibitor of histone deacetylase (HDAC) significantly increased caspase-8 expression in SCLC cells at the RNA and protein levels. Among all studied HDAC inhibitors, valproic acid (VPA) and CI-994 showed prolonged effects on histone acetylation, while combination with decitabine produced the most prominent effects on caspase-8 re-expression. Moreover, a significant reduction of survivin and cIAP-1 proteins level was observed after treatment with VPA. The combination of two drugs sensitized SCLC cells to TRAIL-induced apoptosis, involving mitochondrial apoptotic pathway and was accompanied by Bid cleavage, activation of Bax, and release of cytochrome c. Both initiator caspase-8 and -9 were required for the sensitization of SCLC cells to TRAIL. Thus, efficient restoration of caspase-8 expression in SCLC cells is achieved when a combination of DNMT and HDAC inhibitors is used, suggesting a combination of decitabine and VPA or CI-994 as a potential treatment for sensitization of SCLC cells lacking caspase-8 to TRAIL.
Collapse
Affiliation(s)
- Vitaliy O Kaminskyy
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | | |
Collapse
|
42
|
Lakomy D, Janikashvili N, Fraszczak J, Trad M, Audia S, Samson M, Ciudad M, Vinit J, Vergely C, Caillot D, Foucher P, Lagrost L, Chouaib S, Katsanis E, Larmonier N, Bonnotte B. Cytotoxic dendritic cells generated from cancer patients. THE JOURNAL OF IMMUNOLOGY 2011; 187:2775-82. [PMID: 21804019 DOI: 10.4049/jimmunol.1004146] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Known for years as professional APCs, dendritic cells (DCs) are also endowed with tumoricidal activity. This dual role of DC as killers and messengers may have important implications for tumor immunotherapy. However, the tumoricidal activity of DCs has mainly been investigated in animal models. Cancer cells inhibit antitumor immune responses using numerous mechanisms, including the induction of immunosuppressive/ tolerogenic DCs that have lost their ability to present Ags in an immunogenic manner. In this study, we evaluated the possibility of generating tumor killer DCs from patients with advanced-stage cancers. We demonstrate that human monocyte-derived DCs are endowed with significant cytotoxic activity against tumor cells following activation with LPS. The mechanism of DC-mediated tumor cell killing primarily involves peroxynitrites. This observed cytotoxic activity is restricted to immature DCs. Additionally, after killing, these cytotoxic DCs are able to activate tumor Ag-specific T cells. These observations may open important new perspectives for the use of autologous cytotoxic DCs in cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Daniela Lakomy
- INSERM Unité Mixte de Recherche 866, Institut de Recherche Fédératif 100, Faculté de Médecine, 21079 Dijon, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Fu Q, Wu Y, Yan F, Wang N, Wang W, Cao X, Wang Y, Wan T. Efficient induction of a Her2-specific anti-tumor response by dendritic cells pulsed with a Hsp70L1-Her2(341-456) fusion protein. Cell Mol Immunol 2011; 8:424-32. [PMID: 21785448 DOI: 10.1038/cmi.2011.21] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Heat shock proteins (HSPs) have been shown to interact with antigen-presenting cells (APCs), especially dendritic cells (DCs). HSPs act as potent adjuvants, inducing a Th1 response, as well as antigen-specific CD8(+) cytotoxic T lymphocytes (CTL) via cross-presentation. Our previous work has demonstrated that Hsp70-like protein 1 (Hsp70L1), a new member of the Hsp70 subfamily, can act as a powerful Th1 adjuvant in a DC-based vaccine. Here we report the efficient induction of tumor antigen-specific T cell immune response by DCs pulsed with recombinant fusion protein of Hsp70L1 and Her2(341-456), the latter of which is a fragment of Her2/neu (Her2) containing E75 (a HLA-A2 restricted CTL epitope). The fusion protein Hsp70L1-Her2(341-456) promotes the maturation of DCs and activates them to produce cytokines, such as IL-12 and TNF-α, and chemokines, such as MIP-1α, MIP-1β and RANTES. Taken together, these results indicate that the adjuvant activity of Hsp70L1 is maintained in the fusion protein. Her2-specific HLA-A2.1-restricted CD8(+) CTLs can be generated efficiently either from the Peripheral blood lymphocytes (PBL) of healthy donors or from the splenocytes of immunized HLA-A2.1/K(b) transgenic mice by in vitro stimulation or immunization with DCs pulsed with the Hsp70L1-Her2(341-456) fusion protein. This results in more potent target cell killing in an antigen-specific and HLA-A2.1-restricted manner. Adoptive transfer of splenocytes from transgenic mice immunized with Hsp70L1-Her2(341-456)-pulsed DCs can markedly inhibit tumor growth and prolong the survival of nude mice with Her2(+)/HLA-A2.1(+) human carcinomas. These results suggest that Hsp70L1-Her2(341-456)-pulsed DCs could be a new therapeutic vaccine for patients with Her2(+) cancer.
Collapse
Affiliation(s)
- Qiang Fu
- Department of Oncology, Changhai Hospital of Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Iwami K, Natsume A, Wakabayashi T. Cytokine networks in glioma. Neurosurg Rev 2011; 34:253-63; discussion 263-4. [DOI: 10.1007/s10143-011-0320-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Revised: 12/18/2010] [Accepted: 01/28/2011] [Indexed: 12/25/2022]
|
45
|
Abstract
IMPORTANCE OF THE FIELD TNF-related apoptosis-inducing ligand (TRAIL) is a member of the TNF family of cytokines, which can induce apoptotic cell death in a variety of tumor cells by engaging specific death receptors, TRAIL-R1 and TRAIL-R2, while having low toxicity towards normal cells. There is interest in cancer therapy inducing cell death by activation of the death-receptor-mediated apoptotic pathway while avoiding decoy-receptor-mediated neutralization of the signal. This has led to the development of a number of receptor-specific TRAIL-variants and agonistic antibodies. Some of these soluble recombinant TRAIL and agonist antibodies targeting TRAIL-R1 and/or TRAIL-R2 are progressing in clinical trials. In addition, TRAIL-resistant tumors can be sensitized to TRAIL by a combination of TRAIL or agonistic antibodies with chemotherapeutic agents, targeted small molecules or irradiation. AREAS COVERED IN THIS REVIEW Recent advances in developing TRAIL or its agonist receptor antibodies in cancer therapy. We also discuss combination therapies in overcoming TRAIL resistance in cancer cells. WHAT THE READER WILL GAIN Knowledge of current clinical trials, the promise and obstacles in the future development of therapies affecting TRAIL signaling pathways. TAKE HOME MESSAGE Cancer therapeutics targeting the TRAIL/TRAIL receptor signaling pathway hold great promise for molecularly targeted pro-apoptotic anti-cancer therapy.
Collapse
Affiliation(s)
- Junaid Abdulghani
- Penn State Hershey Medical Center, Penn State Hershey Cancer Institute, Penn State College of Medicine, Department of Medicine (Hematology/Oncology), Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Hershey, PA 17033, USA
| | | |
Collapse
|
46
|
Abstract
Interferon-α (IFN-α), a type I IFN, is a well-known antitumoral agent. The investigation of its clinical properties in acute myeloid leukemia (AML) has been prompted by its pleiotropic antiproliferative and immune effects. So far, integration of IFN-α in the therapeutic arsenal against AML has been modest in view of the divergent results of clinical trials. Recent insights into the key pharmacokinetic determinants of the clinical efficacy of IFN along with advances in its pharmaceutical formulation, have sparked renewed interest in its use. This paper reviews the possible applicability of IFN-α in the treatment of AML and provides a rational basis to re-explore its efficacy in clinical trials.
Collapse
|
47
|
Min Y, Han D, Fu Z, Wang H, Liu L, Tian Y. α-MSH inhibits TNF-α-induced maturation of human dendritic cells in vitro through the up-regulation of ANXA1. Acta Biochim Biophys Sin (Shanghai) 2011; 43:61-8. [PMID: 21131366 DOI: 10.1093/abbs/gmq109] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
α-Melanocyte-stimulating hormone (α-MSH), an anti-inflammatory and immunomodulatory neuropeptide, has been shown to be effective in the experimental treatment of autoimmune diseases and allograft rejection. However, its regulatory mechanism is still unclear. Mature dendritic cells (DCs) are pivotal initiators of immune response and inflammation. We hypothesized that the regulatory role of α-MSH in DC maturation would contribute to the effects of α-MSH in immune-response-mediated disease models. It was found that α-MSH inhibited tumor necrosis factor-alpha (TNF-α)-induced maturation of human peripheral-monocyte-derived DCs (MoDCs), both phenotypically and functionally. This occurred through the down-regulation of the expression of co-stimulatory molecules CD83 and CD86, the production of IL-12, the promotion of IL-10 secretion, and the MoDC phagocytic activity, suggesting that the inhibition of DC maturation by α-MSH could contribute to the anti-inflammatory effect of this neuropeptide. Furthermore, increased expression of annexin A1 (ANXA1) was found to be responsible for the α-MSH inhibiting effect on TNF-α-induced MoDC maturation, which could be abolished by the treatment of MoDCs with specific, small interfering RNAs targeting ANXA1 (ANXA1-siRNA), suggesting that α-MSH-induced ANXA1 mediates the inhibition. Therefore, α-MSH inhibits TNF-α-induced maturation of human DCs through α-MSH-up-regulated ANXA1, suggesting that inhibition of the maturation of DCs by α-MSH could mediate the anti-inflammatory effect of the neuropeptide. Furthermore, ANXA1 could be identified as a new therapeutic drug target based on the role of DCs in immune-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Yan Min
- Institute of Immunology and National Key Laboratory of Medical Immunology, Second Military Medical University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
48
|
Nakamoto Y, Mizukoshi E, Kitahara M, Arihara F, Sakai Y, Kakinoki K, Fujita Y, Marukawa Y, Arai K, Yamashita T, Mukaida N, Matsushima K, Matsui O, Kaneko S. Prolonged recurrence-free survival following OK432-stimulated dendritic cell transfer into hepatocellular carcinoma during transarterial embolization. Clin Exp Immunol 2010; 163:165-77. [PMID: 21087443 DOI: 10.1111/j.1365-2249.2010.04246.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Despite curative locoregional treatments for hepatocellular carcinoma (HCC), tumour recurrence rates remain high. The current study was designed to assess the safety and bioactivity of infusion of dendritic cells (DCs) stimulated with OK432, a streptococcus-derived anti-cancer immunotherapeutic agent, into tumour tissues following transcatheter hepatic arterial embolization (TAE) treatment in patients with HCC. DCs were derived from peripheral blood monocytes of patients with hepatitis C virus-related cirrhosis and HCC in the presence of interleukin (IL)-4 and granulocyte-macrophage colony-stimulating factor and stimulated with 0·1 KE/ml OK432 for 2 days. Thirteen patients were administered with 5 × 10⁶ of DCs through arterial catheter during the procedures of TAE treatment on day 7. The immunomodulatory effects and clinical responses were evaluated in comparison with a group of 22 historical controls treated with TAE but without DC transfer. OK432 stimulation of immature DCs promoted their maturation towards cells with activated phenotypes, high expression of a homing receptor, fairly well-preserved phagocytic capacity, greatly enhanced cytokine production and effective tumoricidal activity. Administration of OK432-stimulated DCs to patients was found to be feasible and safe. Kaplan-Meier analysis revealed prolonged recurrence-free survival of patients treated in this manner compared with the historical controls (P = 0·046, log-rank test). The bioactivity of the transferred DCs was reflected in higher serum concentrations of the cytokines IL-9, IL-15 and tumour necrosis factor-α and the chemokines CCL4 and CCL11. Collectively, this study suggests that a DC-based, active immunotherapeutic strategy in combination with locoregional treatments exerts beneficial anti-tumour effects against liver cancer.
Collapse
Affiliation(s)
- Y Nakamoto
- Cancer Research Institute, Kanazawa University, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
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
|
50
|
Liabeuf S, Barreto DV, Barreto FC, Chasseraud M, Brazier M, Choukroun G, Kamel S, Massy ZA. The circulating soluble TRAIL is a negative marker for inflammation inversely associated with the mortality risk in chronic kidney disease patients. Nephrol Dial Transplant 2010; 25:2596-602. [PMID: 20190248 DOI: 10.1093/ndt/gfq042] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is associated with accelerated atherosclerosis and an inadequate inflammatory response which may account for the high morbidity and mortality observed in this population. In vitro and preclinical evidence suggests that the tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) might be involved in both the atherosclerosis pathway and modulation of the inflammatory response. The aim of the present study was thus to (i) determine serum levels of soluble TRAIL (sTRAIL) in a cohort of CKD patients, (ii) assess the relationship between sTRAIL and other inflammatory biomarkers (C-reactive protein and albumin) and (iii) evaluate the association between serum sTRAIL levels and the mortality risk. METHODS One hundred and thirty patients (mean +/- SD age: 67 +/- 12; 62% males; 8% at CKD stage 2, 26% at stage 3, 27% at stage 4, 8% at stage 5 and 31% at stage 5D) were assayed for sTRAIL and the selected biochemical parameters and then prospectively monitored for mortality. RESULTS CKD stage 5D patients had significantly lower serum sTRAIL levels (median: 46 pg/ml) than patients at CKD stages 2 and 3 (median: 62 pg/ml) or stages 4 and 5 (median: 71 pg/ml). There was no correlation between serum sTRAIL and the estimated glomerular filtration rate (GFR) (r(2) = 0.017, P = 0.22) in pre-dialysis patients. In a multivariate regression analysis, the body mass index (beta = 1.48, P = 0.001) and the serum C-reactive protein (CRP) level (beta = -8.841, P < 0.0001) were independently associated with serum sTRAIL. During follow-up (mean: 772 +/- 286 days), 36 patients died (19 from cardiovascular events, 8 from infectious events and 9 from other causes). The lowest sTRAIL levels (first tertile) were associated with the worst all-cause survival (P = 0.010). Cox regression analyses (with non-cumulative models including age, albumin and CRP as covariates) confirmed the low serum sTRAIL level (first tertile) as an independent predictor of all-cause mortality. CONCLUSIONS Circulating sTRAIL is a negative marker for inflammation and is inversely associated with the mortality risk in CKD patients. Further studies are needed to better understand the role of sTRAIL as an inflammatory marker and to confirm its protective role in the CKD population.
Collapse
|