1
|
Xia M, Hu X, Zhao Q, Ru Y, Wang H, Zheng F. Development and Characterization of a Nanobody against Human T-Cell Immunoglobulin and Mucin-3. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:2929605. [PMID: 35726228 PMCID: PMC9206550 DOI: 10.1155/2022/2929605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/07/2022] [Accepted: 04/19/2022] [Indexed: 11/17/2022]
Abstract
Monoclonal antibodies and antibody-derived biologics are essential tools for cancer research and therapy. The development of monoclonal antibody treatments for successful tumor-targeted therapies took several decades. A nanobody constructed by molecular engineering of heavy-chain-only antibody, which is unique in camel or alpaca, is a burgeoning tools of diagnostic and therapeutic in clinic. In this study, we immunized a 4-year-old female alpaca with TIM-3 antigen. Then, a VHH phage was synthesized from the transcriptome of its B cells by nested PCR as an intermediate library; the library selection for Tim-3 antigen is carried out in three rounds of translation. The most reactive colonies were selected by periplasmic extract monoclonal ELISA. The nanobody was immobilized by metal affinity chromatography (IMAC) purification with the use of a Ni-NTA column, SDS-PAGE, and Western blotting. Finally, the affinity of TIM3-specific nanobody was determined by ELISA. As results, specific 15 kD bands representing nanomaterials were observed on the gel and confirmed by Western blotting. The nanobody showed obvious specific immune response to Tim-3 and had high binding affinity. We have successfully prepared a functional anti-human Tim-3 nanobody with high affinity in vitro.
Collapse
Affiliation(s)
- Mingyuan Xia
- Department of Urology, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an City, 710032 Shaanxi Province, China
| | - Xiangnan Hu
- No. 986 Hospital, Air Force Military Medical University, Xi'an City, 710054 Shaanxi Province, China
| | - Qiuxiang Zhao
- Department of Urology, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an City, 710032 Shaanxi Province, China
| | - Yi Ru
- Department of Biochemistry and Molecular Biology, Basic Medical College, Air Force Military Medical University, Xi'an City, 710032 Shaanxi Province, China
| | - He Wang
- Department of Urology, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an City, 710032 Shaanxi Province, China
| | - Fang Zheng
- The Key Laboratory of Environment and Genes Related to Disease of Ministry of Education, Health Science Center, Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
2
|
Zargar Balajam N, Shabani M, Aghaei M. Galectin-9 inhibits cell proliferation and induces apoptosis in Jurkat and KE-37 acute lymphoblastic leukemia cell lines via caspase-3 activation. Res Pharm Sci 2021; 16:612-622. [PMID: 34760009 PMCID: PMC8562407 DOI: 10.4103/1735-5362.327507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 04/06/2021] [Accepted: 09/12/2021] [Indexed: 11/27/2022] Open
Abstract
Background and purpose: Acute lymphoblastic leukemia (ALL) is a type of cancer of blood and bone marrow characterized by abnormal proliferation of lymphoid progenitor cells. Galectin-9 is a tandem-repeat type galectin expressed in various tumor cells. It seems that the connection between galectin-9 and T cell immunoglobulin mucin-3 receptor acts as a negative regulator of cancer cells proliferation. Experimental approach: In this research, the effects of galectin-9 were investigated using MTS cell proliferation colorimetric, colony-forming, annexin V-FITC/PI, and caspase-3 assays in the Jurkat and KE-37 cell lines of ALL. Furthermore, the western blotting technique was used to evaluate the levels of apoptotic proteins such as Bax and Bcl-2 in these cell lines. Findings/Results: Our results indicated that galectin-9 can considerably reduce the cell growth and colony formation ability of both Jurkat and KE-37 cell lines in a concentration-dependent manner. Besides, galectin-9 induced apoptosis in a concentration-dependent manner in ALL cells by a mechanism associated with Bax/Bcl-2 expression and activation of the caspase-3 activation. Conclusion and implications: Galectin-9 inhibited the growth and proliferation of cell lines with increased programmed cell death, therefore it can be considered as a potential factor in the progression of ALL therapeutics that needs more research in this context.
Collapse
Affiliation(s)
- Narges Zargar Balajam
- Department of Clinical Biochemistry and Isfahan Pharmaceutical Sciences Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Mahdi Shabani
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, I.R. Iran
| | - Mahmoud Aghaei
- Department of Clinical Biochemistry and Isfahan Pharmaceutical Sciences Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| |
Collapse
|
3
|
Guo J, Muse E, Christians AJ, Swanson SJ, Davila E. An Anticancer Drug Cocktail of Three Kinase Inhibitors Improved Response to a Dendritic Cell-Based Cancer Vaccine. Cancer Immunol Res 2019; 7:1523-1534. [PMID: 31266784 PMCID: PMC6726569 DOI: 10.1158/2326-6066.cir-18-0684] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 03/22/2019] [Accepted: 06/28/2019] [Indexed: 12/17/2022]
Abstract
Monocyte-derived dendritic cell (moDC)-based cancer therapies intended to elicit antitumor T-cell responses have limited efficacy in most clinical trials. However, potent and sustained antitumor activity in a limited number of patients highlights the therapeutic potential of moDCs. In vitro culture conditions used to generate moDCs can be inconsistent, and moDCs generated in vitro are less effective than natural DCs. On the basis of our study highlighting the ability for certain kinase inhibitors to enhance tumor antigenicity, we therefore screened kinase inhibitors for their ability to improve DC immunogenicity. We identified AKT inhibitor MK2206, DNA-PK inhibitor NU7441, and MEK inhibitor trametinib as the compounds most effective at modulating moDC immunogenicity. The combination of these drugs, referred to as MKNUTRA, enhanced moDC activity over treatment with individual drugs while exhibiting minimal toxicity. An evaluation of 335 activation and T-cell-suppressive surface proteins on moDCs revealed that MKNUTRA treatment more effectively matured cells and reduced the expression of tolerogenic proteins as compared with control moDCs. MKNUTRA treatment imparted to ICT107, a glioblastoma (GBM) DC-based vaccine that has completed phase II trials, an increased ability to stimulate patient-derived autologous CD8+ T cells against the brain tumor antigens IL13Rα2(345-354) and TRP2(180-188) In vivo, treating ICT107 with MKNUTRA, prior to injection into mice with an established GBM tumor, reduced tumor growth kinetics. This response was associated with an increased frequency of tumor-reactive lymphocytes within tumors and in peripheral tissues. These studies broaden the application of targeted anticancer drugs and highlight their ability to increase moDC immunogenicity.
Collapse
Affiliation(s)
- Jitao Guo
- Division of Medical Oncology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Elena Muse
- Division of Medical Oncology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Allison J Christians
- Division of Medical Oncology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | | | - Eduardo Davila
- Division of Medical Oncology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
- Human Immunology and Immunotherapy Initiative, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Comprehensive Cancer Center, Aurora, Colorado
| |
Collapse
|
4
|
唐 映, 徐 加. Tim-3在肝脏疾病中的调节作用. Shijie Huaren Xiaohua Zazhi 2017; 25:2080-2087. [DOI: 10.11569/wcjd.v25.i23.2080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
T淋巴细胞免疫球蛋白黏蛋白分子(T-cell immunoglobulin domain and mucin domain-containing molecule, Tim)-3是Tim家族中的一员, 为近年来新发现的一种在辅助Ⅰ型T淋巴细胞(Help T cell 1, Th1)上特异性表达的Ⅰ型细胞表面分子. Tim-3作为负性调节因子通过与其配体Galectin-9结合引起细胞死亡, 进而调控Th1型细胞功能. Tim-3还表达于其他类型细胞表面, 如自然杀伤细胞、树突状细胞和单核细胞, 对自身免疫性疾病和其他免疫介导的疾病进行免疫调控. 对Tim-3在不同细胞不同免疫条件下的功能以及如何调节进行研究, 将有利于研发Tim-3的潜在治疗作用. 近年来大量研究显示Tim-3通道与肝脏疾病发生发展有着密切关系, 本文就其在肝脏疾病中的调节作用做一总结.
Collapse
|
5
|
Li G, Wang Z, Zhang C, Liu X, Cai J, Wang Z, Hu H, Wu F, Bao Z, Liu Y, Zhao L, Liang T, Yang F, Huang R, Zhang W, Jiang T. Molecular and clinical characterization of TIM-3 in glioma through 1,024 samples. Oncoimmunology 2017; 6:e1328339. [PMID: 28919992 DOI: 10.1080/2162402x.2017.1328339] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/02/2017] [Accepted: 05/05/2017] [Indexed: 10/19/2022] Open
Abstract
Background: Researches on immunotherapy of glioma has been increasing exponentially in recent years. However, autoimmune-like side effects of current immune checkpoint blockade hindered the clinical application of immunotherapy in glioma. The discovery of the TIM-3, a tumor-specific immune checkpoint, has shed a new light on solution of this dilemma. We aimed at investigating the role of TIM-3 at transcriptome level and its relationship with clinical practice in glioma. Methods: A cohort of 325 glioma patients with RNA-seq data from Chinese Glioma Genome Atlas (CGGA project) was analyzed, and the results were well validated in TCGA RNA-seq data of 699 gliomas. R language was used as the main tool for statistical analysis and graphical work. Results: TIM-3 was enriched in glioblastoma (the most malignant glioma) and IDH-wildtype glioma. TIM-3 can act as a potential marker for mesenchymal molecular subtype according to TCGA transcriptional classification scheme in glioma. TIM-3 was closely related to immune functions in glioma, especially T cell mediated immune response to tumor cell and T cell mediated cytotoxicity directed against tumor cell target. Moreover, TIM-3 and PD-L1 played almost exactly the same inflammatory activation functions in glioma. Clinically, high expression of TIM-3 was an independent indicator of poor prognosis. Conclusion: The expression of TIM-3 is closely related to the pathology and molecular pathology of glioma. Meanwhile, in glioma TIM-3 plays a specific role in T cell tumor immune response. Therefore, TIM-3 is a promising target for immunotherapeutic strategies, providing an alternative treatment when glioma gains resistance to antibodies of PD-1/PD-L1.
Collapse
Affiliation(s)
- Guanzhang Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA)
| | - Zheng Wang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA)
| | - Chuanbao Zhang
- Chinese Glioma Genome Atlas Network (CGGA).,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xing Liu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA)
| | - Jinquan Cai
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhiliang Wang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA)
| | - Huimin Hu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA)
| | - Fan Wu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA)
| | - Zhaoshi Bao
- Chinese Glioma Genome Atlas Network (CGGA).,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yanwei Liu
- Chinese Glioma Genome Atlas Network (CGGA).,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Liang Zhao
- Department of Neurosurgery, University Medical Center Düsseldorf, Düsseldorf, Germany
| | - Tingyu Liang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA)
| | - Fan Yang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA)
| | - Ruoyu Huang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA)
| | - Wei Zhang
- Chinese Glioma Genome Atlas Network (CGGA).,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tao Jiang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA).,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Institute for Brain Disorders Brain Tumor Center, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
6
|
Xu G, Zheng K, Lu X, Wang J, Chai Y, Wang J. Association between polymorphisms in the promoter region of T cell immunoglobulin and mucin domain-3 and myasthenia gravis-associated thymoma. Oncol Lett 2015; 9:1470-1474. [PMID: 25663933 PMCID: PMC4314971 DOI: 10.3892/ol.2015.2845] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 12/12/2014] [Indexed: 11/06/2022] Open
Abstract
Thymoma is a type of benign or low-grade malignant tumor, occurring on the thymic epithelium. Patients with thymoma may also suffer from myasthenia gravis (MG), presenting MG-associated thymoma. T cell immunoglobulin and mucin domain-3 (Tim-3), a subtype of the Tim protein family, may be an important immune regulatory and pivotal molecule associated with tumor development. In order to understand the etiology and pathogenesis of MG-associated thymoma in the Han population of North China, the present study investigated the association between a polymorphism on the -574 locus in the promoter of Tim-3 and the risk of MG-associated thymoma in the Han Chinese population. In total, 116 patients with thymoma and MG were enrolled into the MG-associated thymoma group, while 124 patients with thymoma, but without MG, were enrolled into the non-MG-associated thymoma group. Examinations were conducted to reach a definite diagnosis of thymoma and MG and rule out other autoimmune diseases. Allele-specific polymerase chain reaction (AS-PCR) was performed to determine the polymorphism on the -574 locus of Tim-3 in all the subjects. PCR products were randomly selected for sequencing. Statistically significant differences were detected between the distribution frequencies of the GT+TT genotype and T allele on the -574 locus of the MG-associated thymoma group (31.03 vs. 12.90%, respectively; χ2=11.609, P=0.001) and the non-MG-associated thymoma group (15.52 vs. 6.45%, respectively; χ2=10.198, P=0.001). In conclusion, the present study indicated that an association may exist between the polymorphism of the -574 locus in the Tim-3 promoter and MG-associated thymoma.
Collapse
Affiliation(s)
- Guowu Xu
- Department of Emergency, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Kai Zheng
- Department of Cardiothoracic Surgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Xing Lu
- Department of Intensive Care Unit, Tianjin Third Central Hospital, Tianjin 300170, P.R. China
| | - Jinxiang Wang
- Department of Emergency, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yanfen Chai
- Department of Emergency, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Junyi Wang
- Department of Intensive Care Unit, Tianjin Third Central Hospital, Tianjin 300170, P.R. China
| |
Collapse
|
7
|
Baghdadi M, Jinushi M. The impact of the TIM gene family on tumor immunity and immunosuppression. Cell Mol Immunol 2014; 11:41-8. [PMID: 24336162 PMCID: PMC4002149 DOI: 10.1038/cmi.2013.57] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 10/16/2013] [Accepted: 10/17/2013] [Indexed: 12/13/2022] Open
Abstract
Tumor immunoevasion is an advanced phase of cancer immunosurveillance in which tumor cells acquire the ability to circumvent host immune systems and exploit protumorigenic inflammation. T-cell immunoglobulin mucin (TIM) gene family members have emerged as critical checkpoint proteins that regulate multiple immune response phases and maintain immune homeostasis. Accumulating evidence demonstrates that tumor cells exploit TIM gene family members to evade immunosurveillance, whereas TIM gene family members facilitate the prevention of inflammation-related tumor progression. Thus, a comprehensive analysis to clarify the relative contributions of TIM gene family members in tumor progression may elucidate immunosurveillance systems in cancer patients.
Collapse
Affiliation(s)
| | - Masahisa Jinushi
- Research Center for Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Hokkaido, Japan
| |
Collapse
|
8
|
Schiavoni G, Gabriele L, Mattei F. The tumor microenvironment: a pitch for multiple players. Front Oncol 2013; 3:90. [PMID: 23616948 PMCID: PMC3628362 DOI: 10.3389/fonc.2013.00090] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 04/03/2013] [Indexed: 12/13/2022] Open
Abstract
The cancer microenvironment may be conceptually regarded as a pitch where the main players are resident and non-resident cellular components, each covering a defined role and interconnected by a complex network of soluble mediators. The crosstalk between these cells and the tumor cells within this environment crucially determines the fate of tumor progression. Immune cells that infiltrate the tumor bed are transported there by blood circulation and exert a variety of effects, either counteracting or favoring tumor outgrowth. Here, we review and discuss the multiple populations composing the tumor bed, with special focus on immune cells subsets that positively or negatively dictate neoplastic progression. In this scenario, the contribution of cancer stem cells within the tumor microenvironment will also be discussed. Finally, we illustrate recent advances on new integrated approaches to investigate the tumor microenvironment in vitro.
Collapse
Affiliation(s)
- Giovanna Schiavoni
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità Rome, Italy
| | | | | |
Collapse
|