1
|
Du X, Guo Y, Zhao X, Zhang L, Fan R, Li Y. METTL3-mediated TIM1 promotes macrophage M1 polarization and inflammation through IGF2BP2-dependent manner. J Biochem Mol Toxicol 2024; 38:e23845. [PMID: 39267336 DOI: 10.1002/jbt.23845] [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: 04/25/2024] [Revised: 07/22/2024] [Accepted: 08/29/2024] [Indexed: 09/17/2024]
Abstract
Macrophage polarization and inflammation may play an important role in the development of sepsis. T-cell immunoglobulin mucin 1 (TIM1) has been demonstrated to promote macrophage inflammatory responses. However, whether TIM1 regulates macrophage polarization and inflammation to affect sepsis development remains unclear. Human monocytic leukemia cell line was induced into macrophages, followed by stimulated with LPS and IL-4 to induce M1 polarization and M2 polarization. The expression levels of TIM1, methyltransferase 3 (METTL3), and insulin like growth factor 2 mRNA binding protein 2 (IGF2BP2) were examined by qRT-PCR and western blot. IL-6, IL-1β, and TNF-α levels were tested by ELISA. CD86+cell rate was analyzed by flow cytometry. The m6A methylation level of TIM1 was assessed by MeRIP assay. The interaction of between TIM1 and METTL3 or IGF2BP2 was assessed by dual-luciferase reporter assay and RIP assay. TIM1 knockdown repressed LPS-induced macrophage M1 polarization and inflammation. In terms of mechanism, METTL3 promoted TIM1 expression through m6A modification, and this modification could be recognized by IGF2BP2. Besides, knockdown of METTL3/IGF2BP2 suppressed LPS-induced macrophage M1 polarization and inflammation, while this effect could be eliminated by TIM1 overexpression. METTL3/IGF2BP2/TIM1 axis promoted macrophage M1 polarization and inflammation, which might provide potential target for sepsis treatment.
Collapse
Affiliation(s)
- Xianrong Du
- The Geriatrics Department of Shanxi Provincial People Hospital, Shanxi Medical University, Taiyuan, China
| | - Yinguang Guo
- Physical Examination Center of Shanxi Provincial Hospital of Traditional Chinese Medicine, Taiyuan, China
| | - Xiaoqin Zhao
- School of Physical Education, Taiyuan University of Technology, Taiyuan, China
| | - Lijuan Zhang
- The Nephrology Department of Shanxi Provincial People Hospital, Shanxi Medical University, Taiyuan, China
| | - Ru Fan
- The Nephrology Department of Shanxi Provincial People Hospital, Shanxi Medical University, Taiyuan, China
| | - Yafeng Li
- Core Laboratory of Shanxi Provincial People Hospital, Shanxi Medical University, Taiyuan, China
- The Nephrology Department of Hejin Municipal People Hospital, Yuncheng, China
- Academy of Microbial Ecology, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
2
|
Lin L, Zhao Y, Ma Y, Xi K, Jin Y, Huang X, Huang Y, Zhang Y, Qin Q. Grouper TIM-1 promotes nodavirus infection by inhibiting immune and inflammation response. FISH & SHELLFISH IMMUNOLOGY 2024; 153:109822. [PMID: 39117128 DOI: 10.1016/j.fsi.2024.109822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 08/10/2024]
Abstract
T-cell/transmembrane immunoglobulin and mucin domain-containing (TIM) protein family has attracted particular attention because of their broad immune functions and the response to viral infections. TIM-1, a member of the TIM family, has been demonstrated to play an important role in viral infections. However, its roles during fish nodavirus infection still remained largely unknown. In this study, a homolog of TIM-1 from orange-spotted grouper (Epinephelus coioides) (EcTIM-1) was identified, and characterized. EcTIM-1 encoded a 217-amino acids protein, containing one Immunoglobulin domain. Homology analysis showed that EcTIM-1 shared 98.62 % and 42.99 % identity to giant grouper (E. lanceolatus) and human (Homo sapiens). Quantitative Real-time PCR analyses indicated that EcTIM-1 was expressed in all examined tissues, with higher expression in liver, spleen, skin, and heart, and was significantly up-regulated in response to red-spotted grouper nervous necrosis virus (RGNNV) infection. EcTIM-1 was distributed in the cytoplasm, and partly co-localized with Golgi apparatus and lysosomes in vitro. The ectopic expression of EcTIM-1 promoted RGNNV replication by increasing the level of viral genes transcription and protein synthesis. Besides, overexpression of EcTIM-1 decreased the luciferase activity of type I interferon (IFN1), interferon stimulated response elements (ISRE) and nuclear factor kappa-B (NF-κB) promoters, as well as the transcription of pro-inflammatory factors and interferon related genes. EcTIM-1 significantly suppressed the luciferase activity of IFN1, ISRE and NF-κB promoters evoked by Epinephelus coioides melanoma differentiation-associated gene 5 (EcMDA5), mitochondrial antiviral signaling protein (EcMAVS), stimulator of IFN genes (EcSTING) or TANK-binding kinase 1 (EcTBK1). Collectively, EcTIM-1 negatively regulated interferon and inflammatory response to promote RGNNV infection. These results provide a basis for a better understanding of the innate immune response of TIM-1 in fish.
Collapse
Affiliation(s)
- Long Lin
- College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Yin Zhao
- College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Yiting Ma
- College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Kaitao Xi
- College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Yunyong Jin
- College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Xiaohong Huang
- College of Marine Sciences, South China Agricultural University, Guangzhou, China; Nansha-South China Agricultural University Fishery Research Institute, Guangzhou, China
| | - Youhua Huang
- College of Marine Sciences, South China Agricultural University, Guangzhou, China; Nansha-South China Agricultural University Fishery Research Institute, Guangzhou, China
| | - Ya Zhang
- College of Marine Sciences, South China Agricultural University, Guangzhou, China; Nansha-South China Agricultural University Fishery Research Institute, Guangzhou, China.
| | - Qiwei Qin
- College of Marine Sciences, South China Agricultural University, Guangzhou, China; Nansha-South China Agricultural University Fishery Research Institute, Guangzhou, China; Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai, China.
| |
Collapse
|
3
|
Breinbauer R, Mäling M, Ehnert S, Blumenstock G, Schwarz T, Jazewitsch J, Erne F, Reumann MK, Rollmann MF, Braun BJ, Histing T, Nüssler AK. B7-1 and PlGF-1 are two possible new biomarkers to identify fracture-associated trauma patients at higher risk of developing complications: a cohort study. BMC Musculoskelet Disord 2024; 25:677. [PMID: 39210389 PMCID: PMC11360573 DOI: 10.1186/s12891-024-07789-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Around 10% of fractures lead to complications. With increasing fracture incidences in recent years, this poses a serious burden on the healthcare system, with increasing costs for treatment. In the present study, we aimed to identify potential 'new' blood markers to predict the development of post-surgical complications in trauma patients following a fracture. METHODS A total of 292 trauma patients with a complete three-month follow-up were included in this cohort study. Blood samples were obtained from 244 of these patients. Two complication groups were distinguished based on the Clavien-Dindo (CD) classification: CD grade I and CD grade III groups were compared to the controls (CD 0). The Mann-Whitney U test was used to compare the complication groups to the control group. RESULTS Analysis of the patients' data revealed that risk factors are dependent on sex. Both, males and females who developed a CD III complication showed elevated blood levels of B7-1 (p = 0.015 and p = 0.018, respectively) and PlGF-1 (p = 0.009 and p = 0.031, respectively), with B7-1 demonstrating greater sensitivity (B7-1: 0.706 (male) and 0.692 (female), PlGF-1: 0.647 (male) and 0.615 (female)). Further analysis of the questionnaires and medical data revealed the importance of additional risk factors. For males (CD 0: 133; CD I: 12; CD III: 18 patients) alcohol consumption was significantly increased for CD I and CD III compared to control with p = 0.009 and p = 0.007, respectively. For females (CD 0: 107; CD I: 10; CD III: 12 patients) a significantly increased average BMI [kg/m2] from 25.5 to 29.7 with CD III was observed, as well as an elevation from one to three comorbidities (p = 0.003). CONCLUSIONS These two potential new blood markers hold promise for predicting complication development in trauma patients. Nevertheless, further studies are necessary to evaluate the diagnostic utility of B7-1 and PlGF-1 in predicting complications in trauma patients and consider sex differences before their possible use as routine clinical screening tools.
Collapse
Affiliation(s)
- Regina Breinbauer
- Siegfried-Weller-Institute, BG Unfallklinik Tuebingen, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
| | - Michelle Mäling
- Siegfried-Weller-Institute, BG Unfallklinik Tuebingen, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
| | - Sabrina Ehnert
- Siegfried-Weller-Institute, BG Unfallklinik Tuebingen, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
| | - Gunnar Blumenstock
- Department of Clinical Epidemiology and Applied Biometry, Eberhard Karls University Tuebingen, Silcherstrasse 5, 72076, Tuebingen, Germany
| | - Tobias Schwarz
- Siegfried-Weller-Institute, BG Unfallklinik Tuebingen, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
| | - Johann Jazewitsch
- Siegfried-Weller-Institute, BG Unfallklinik Tuebingen, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
| | - Felix Erne
- Siegfried-Weller-Institute, BG Unfallklinik Tuebingen, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
- Department of Traumatology and Reconstructive Surgery, BG Unfallklinik Tuebingen, Eberhard Karls University Tuebingen, Schnarrenbergstr. 95, 72076, Tuebingen, Germany
| | - Marie K Reumann
- Siegfried-Weller-Institute, BG Unfallklinik Tuebingen, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
- Department of Traumatology and Reconstructive Surgery, BG Unfallklinik Tuebingen, Eberhard Karls University Tuebingen, Schnarrenbergstr. 95, 72076, Tuebingen, Germany
| | - Mika F Rollmann
- Department of Traumatology and Reconstructive Surgery, BG Unfallklinik Tuebingen, Eberhard Karls University Tuebingen, Schnarrenbergstr. 95, 72076, Tuebingen, Germany
| | - Benedikt J Braun
- Department of Traumatology and Reconstructive Surgery, BG Unfallklinik Tuebingen, Eberhard Karls University Tuebingen, Schnarrenbergstr. 95, 72076, Tuebingen, Germany
| | - Tina Histing
- Department of Traumatology and Reconstructive Surgery, BG Unfallklinik Tuebingen, Eberhard Karls University Tuebingen, Schnarrenbergstr. 95, 72076, Tuebingen, Germany
| | - Andreas K Nüssler
- Siegfried-Weller-Institute, BG Unfallklinik Tuebingen, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany.
| |
Collapse
|
4
|
Biały S, Siemaszko J, Sobczyk-Kruszelnicka M, Fidyk W, Solarska I, Nasiłowska-Adamska B, Skowrońska P, Bieniaszewska M, Tomaszewska A, Basak GW, Giebel S, Wróbel T, Bogunia-Kubik K. Unravelling the potential of TIM-3 gene polymorphism in allogeneic hematopoietic stem cell transplantation - a preliminary study. Transpl Immunol 2024; 85:102084. [PMID: 38992477 DOI: 10.1016/j.trim.2024.102084] [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: 06/04/2024] [Revised: 07/01/2024] [Accepted: 07/07/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) molecule is a key regulator of the immune response by exerting an inhibitory effect on various types of immune cells. Understanding the role of TIM-3 in hematopoietic stem cell transplantation (HSCT) may improve transplant outcomes. Our study evaluated the potential association between TIM-3 polymorphisms, namely rs1036199 (A > C) or rs10515746 (C > A), changes which are located in exon 3 and the promoter region of the TIM-3 gene, and post-HSCT outcomes. METHODS One-hundred and twenty allogeneic HSCT patients and their respective donors were enrolled and genotyped for TIM-3 single nucleotide polymorphisms (SNPs) using real-time PCR with TaqMan assays. RESULTS We found that the presence of the rare alleles and heterozygous genotypes of studied SNP in recipients tended to protect against or increase the risk for acute graft-versus-host disease (aGvHD). For the rs1036199 polymorphism, recipients with the AC heterozygous genotype (p = 0.0287) or carrying the rarer C allele (p = 0.0334) showed a lower frequency of aGvHD development along all I-IV grades. A similar association was detected for the rs10515746 polymorphism as recipients with the CA genotype (p = 0.0095) or the recessive A allele (p = 0.0117) less frequently developed aGvHD. Furthermore, the rarer A allele of rs10515746 SNP was also associated with a prolonged aGvHD-free survival (p = 0.0424). Cytomegalovirus (CMV) infection was more common in patients transplanted with TIM-3 rs10515746 mismatched donors (p = 0.0229) and this association was also found to be independent of HLA incompatibility and pre-transplant CMV-IgG status. Multivariate analyses confirmed the role of these recessive alleles and donor-recipient TIM-3 incompatibility as an independent factor in aGvHD and CMV development. CONCLUSIONS Polymorphism of TIM-3 molecule may affect the immune response in HSCT patients. The recessive alleles of rs1036199 and rs10515746 SNPs decreased the risk of developing aGvHD. TIM-3 donor-recipient genetic matching may also affect the risk of post-transplant CMV infection, indicating the potential value of genetic profiling in optimizing transplant strategies.
Collapse
Affiliation(s)
- Sylwia Biały
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Jagoda Siemaszko
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Małgorzata Sobczyk-Kruszelnicka
- Department of Bone Marrow Transplantation and Hematology-Oncology, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Wojciech Fidyk
- Department of Bone Marrow Transplantation and Hematology-Oncology, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Iwona Solarska
- Institute of Hematology and Blood Transfusion Medicine, Warsaw, Poland
| | | | | | - Maria Bieniaszewska
- Department of Hematology and Transplantology, Medical University of Gdansk, Gdansk, Poland
| | - Agnieszka Tomaszewska
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Grzegorz W Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Sebastian Giebel
- Department of Bone Marrow Transplantation and Hematology-Oncology, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Tomasz Wróbel
- Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| | - Katarzyna Bogunia-Kubik
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland.
| |
Collapse
|
5
|
Al-Danakh A, Safi M, Jian Y, Yang L, Zhu X, Chen Q, Yang K, Wang S, Zhang J, Yang D. Aging-related biomarker discovery in the era of immune checkpoint inhibitors for cancer patients. Front Immunol 2024; 15:1348189. [PMID: 38590525 PMCID: PMC11000233 DOI: 10.3389/fimmu.2024.1348189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 01/29/2024] [Indexed: 04/10/2024] Open
Abstract
Older patients with cancer, particularly those over 75 years of age, often experience poorer clinical outcomes compared to younger patients. This can be attributed to age-related comorbidities, weakened immune function, and reduced tolerance to treatment-related adverse effects. In the immune checkpoint inhibitors (ICI) era, age has emerged as an influential factor impacting the discovery of predictive biomarkers for ICI treatment. These age-linked changes in the immune system can influence the composition and functionality of tumor-infiltrating immune cells (TIICs) that play a crucial role in the cancer response. Older patients may have lower levels of TIICs infiltration due to age-related immune senescence particularly T cell function, which can limit the effectivity of cancer immunotherapies. Furthermore, age-related immune dysregulation increases the exhaustion of immune cells, characterized by the dysregulation of ICI-related biomarkers and a dampened response to ICI. Our review aims to provide a comprehensive understanding of the mechanisms that contribute to the impact of age on ICI-related biomarkers and ICI response. Understanding these mechanisms will facilitate the development of treatment approaches tailored to elderly individuals with cancer.
Collapse
Affiliation(s)
- Abdullah Al-Danakh
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Mohammed Safi
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yuli Jian
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Linlin Yang
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xinqing Zhu
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Qiwei Chen
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Kangkang Yang
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
| | - Shujing Wang
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Deyong Yang
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Surgery, Healinghands Clinic, Dalian, Liaoning, China
| |
Collapse
|
6
|
Sarkar A, Nagappa M, Dey S, Mondal S, Babu GS, Choudhury SP, Akhil P, Debnath M. Synergistic effects of immune checkpoints and checkpoint inhibitors in inflammatory neuropathies: Implications and mechanisms. J Peripher Nerv Syst 2024; 29:6-16. [PMID: 37988274 DOI: 10.1111/jns.12605] [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: 09/02/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 11/23/2023]
Abstract
Immune checkpoint molecules play pivotal roles in the regulation of immune homeostasis. Disruption of the immune checkpoints causes autoimmune/inflammatory as well as malignant disorders. Over the past few years, the immune checkpoint molecules with inhibitory function emerged as potential therapeutic targets in oncological conditions. The inhibition of the function of these molecules by using immune checkpoint inhibitors (ICIs) has brought paradigmatic changes in cancer therapy due to their remarkable clinical benefits, not only in improving the quality of life but also in prolonging the survival time of cancer patients. Unfortunately, the ICIs soon turned out to be a "double-edged sword" as the use of ICIs caused multiple immune-related adverse effects (irAEs). The development of inflammatory neuropathies such as Guillain-Barré syndrome (GBS) and Chronic Inflammatory Demyelinating Polyradiculoneuropathy (CIDP) as the secondary effects of immunotherapy appeared very challenging as these conditions result in significant and often permanent disability. The underlying mechanism(s) through which ICIs trigger inflammatory neuropathies are currently not known. Compelling evidence suggests autoimmune reaction and/or inflammation as the independent risk mechanism of inflammatory neuropathies. There is a lack of understanding as to whether prior exposure to the risk factors of inflammatory neuropathies, the presence of germline genetic variants in immune function-related genes, genetic variations within immune checkpoint molecules, the existence of autoantibodies, and activated/memory T cells act as determining factors for ICI-induced inflammatory neuropathies. Herein, we highlight the available pieces of evidence, discuss the mechanistic basis, and propose a few testable hypotheses on inflammatory neuropathies as irAEs of immunotherapy.
Collapse
Affiliation(s)
- Aritrani Sarkar
- Department of Human Genetics, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Madhu Nagappa
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Saikat Dey
- Department of Human Genetics, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Sandipan Mondal
- Department of Human Genetics, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Gopika Suresh Babu
- Department of Human Genetics, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Saptamita Pal Choudhury
- Department of Human Genetics, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Pokala Akhil
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Monojit Debnath
- Department of Human Genetics, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| |
Collapse
|
7
|
Ebrahimi N, Abdulwahid AHRR, Mansouri A, Karimi N, Bostani RJ, Beiranvand S, Adelian S, Khorram R, Vafadar R, Hamblin MR, Aref AR. Targeting the NF-κB pathway as a potential regulator of immune checkpoints in cancer immunotherapy. Cell Mol Life Sci 2024; 81:106. [PMID: 38418707 PMCID: PMC10902086 DOI: 10.1007/s00018-023-05098-8] [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: 07/02/2023] [Revised: 10/01/2023] [Accepted: 10/29/2023] [Indexed: 03/02/2024]
Abstract
Advances in cancer immunotherapy over the last decade have led to the development of several agents that affect immune checkpoints. Inhibitory receptors expressed on T cells that negatively regulate the immune response include cytotoxic T‑lymphocyte antigen 4 (CTLA4) and programmed cell death protein 1 (PD1), which have been studied more than similar receptors. Inhibition of these proteins and other immune checkpoints can stimulate the immune system to attack cancer cells, and prevent the tumor from escaping the immune response. However, the administration of anti-PD1 and anti-CTLA4 antibodies has been associated with adverse inflammatory responses similar to autoimmune diseases. The current review discussed the role of the NF-κB pathway as a tumor promoter, and how it can govern inflammatory responses and affect various immune checkpoints. More precise knowledge about the communication between immune checkpoints and NF-κB pathways could increase the effectiveness of immunotherapy and reduce the adverse effects of checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Nasim Ebrahimi
- Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran
| | | | - Atena Mansouri
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Nasrin Karimi
- Department of Biology, Faculty of Basic Science, Islamic Azad University Damghan Branch, Damghan, Iran
| | | | - Sheida Beiranvand
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Samaneh Adelian
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Roya Khorram
- Bone and Joint Diseases Research Center, Department of Orthopedic Surgery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Vafadar
- Department of Orthopeadic Surgery, Kerman University of Medical Sciences, Kerman, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa.
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Amir Reza Aref
- Xsphera Biosciences, Translational Medicine Group, 6 Tide Street, Boston, MA, 02210, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
8
|
Raskova Kafkova L, Mierzwicka JM, Chakraborty P, Jakubec P, Fischer O, Skarda J, Maly P, Raska M. NSCLC: from tumorigenesis, immune checkpoint misuse to current and future targeted therapy. Front Immunol 2024; 15:1342086. [PMID: 38384472 PMCID: PMC10879685 DOI: 10.3389/fimmu.2024.1342086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/17/2024] [Indexed: 02/23/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is largely promoted by a multistep tumorigenesis process involving various genetic and epigenetic alterations, which essentially contribute to the high incidence of mortality among patients with NSCLC. Clinical observations revealed that NSCLC also co-opts a multifaceted immune checkpoint dysregulation as an important driving factor in NSCLC progression and development. For example, a deregulated PI3K/AKT/mTOR pathway has been noticed in 50-70% of NSCLC cases, primarily modulated by mutations in key oncogenes such as ALK, EGFR, KRAS, and others. Additionally, genetic association studies containing patient-specific factors and local reimbursement criteria expose/reveal mutations in EGFR/ALK/ROS/BRAF/KRAS/PD-L1 proteins to determine the suitability of available immunotherapy or tyrosine kinase inhibitor therapy. Thus, the expression of such checkpoints on tumors and immune cells is pivotal in understanding the therapeutic efficacy and has been extensively studied for NSCLC treatments. Therefore, this review summarizes current knowledge in NSCLC tumorigenesis, focusing on its genetic and epigenetic intricacies, immune checkpoint dysregulation, and the evolving landscape of targeted therapies. In the context of current and future therapies, we emphasize the significance of antibodies targeting PD-1/PD-L1 and CTLA-4 interactions as the primary therapeutic strategy for immune system reactivation in NSCLC. Other approaches involving the promising potential of nanobodies, probodies, affibodies, and DARPINs targeting immune checkpoints are also described; these are under active research or clinical trials to mediate immune regulation and reduce cancer progression. This comprehensive review underscores the multifaceted nature, current state and future directions of NSCLC research and treatment.
Collapse
Affiliation(s)
- Leona Raskova Kafkova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czechia
- Department of Immunology, University Hospital Olomouc, Olomouc, Czechia
| | - Joanna M. Mierzwicka
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
| | - Prosenjit Chakraborty
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czechia
| | - Petr Jakubec
- Department of Respiratory Diseases and Tuberculosis, University Hospital Olomouc, Olomouc, Czechia
| | - Ondrej Fischer
- Department of Respiratory Diseases and Tuberculosis, University Hospital Olomouc, Olomouc, Czechia
| | - Jozef Skarda
- Institute of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czechia
- Department of Pathology, University Hospital Ostrava and Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| | - Petr Maly
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
| | - Milan Raska
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czechia
- Department of Immunology, University Hospital Olomouc, Olomouc, Czechia
| |
Collapse
|
9
|
Tian W, Liu M, Liu Y, Lv Q, Cheng H, Gu Y, Li M. TIM-3 regulates the proliferation by BDNF-mediated PI3K/AKT axis in the process of endometriosis. Mol Med 2023; 29:170. [PMID: 38114892 PMCID: PMC10731854 DOI: 10.1186/s10020-023-00768-6] [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: 07/13/2023] [Accepted: 12/08/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND T cell immunoglobulin and mucin domain-containing molecule-3 (TIM-3) initially discovered on the surface of Th1 cells, negatively regulates immune responses and mediates apoptosis of Th1 cells. An increasing number of studies have since shown that TIM-3 is crucial in the genesis and development of immune diseases, cancers, and chronic infectious illnesses. However, the effect of TIM-3 on endometriosis is still unknown. METHODS Quantitative real-time polymerase chain reaction, western blotting, and immunohistochemistry were used to measure TIM-3 levels in endometriosis. Cell Counting Kit-8, 5-ethynyl-2'-deoxyuridine, colony-forming, Transwell® migration, Matrigel® invasion, and flow cytometry assays were used to explore the function of TIM-3 in vitro, and xenograft experiments in nude mice were used to assess its role in vivo. According to the RNA seq, brain-derived neurotrophic factor (BDNF) was screened. The involvement of specific proliferation-related signaling molecules was determined by transfecting a plasmid and adding an inhibitor in vivo and in vitro. RESULTS TIM-3 mRNA and protein expression levels were significantly higher in eutopic and ectopic endometrial tissues than in normal endometrial tissues. By examining the effects of TIM-3 overexpression and knockdown on cell proliferation, migration, and invasion in vitro, and lesions formation in vivo, we found that the expression of TIM-3 was positively correlated with cell proliferation and clone formation in vitro, as well as lesions growth in nude mice. By adding the phosphatidylinositol 3 kinase/protein kinase B(PI3K/AKT) pathway inhibitor LY294002 and knocking down PI3K, we further verified that TIM-3 promotes proliferation in vivo and in vitro via the PI3K pathway. By transfecting the plasmid into ESC cells and gave inhibitors to endometriotic rats models, we tested that TIM-3 regulates the proliferation by BDNF-mediated PI3K/AKT axis. CONCLUSION TIM-3 can promote the proliferation of endometriosis by BDNF-mediated PI3K/AKT axis in vivo and in vitro, which may provide a new therapeutic target for the treatment of endometriosis.
Collapse
Affiliation(s)
- Wei Tian
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| | - Min Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| | - Yuqiu Liu
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Qingfeng Lv
- Department of Obstetrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - HuaFeng Cheng
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yanling Gu
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Mingjiang Li
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
10
|
Ao YQ, Gao J, Wang S, Jiang JH, Deng J, Wang HK, Xu B, Ding JY. Immunotherapy of thymic epithelial tumors: molecular understandings and clinical perspectives. Mol Cancer 2023; 22:70. [PMID: 37055838 PMCID: PMC10099901 DOI: 10.1186/s12943-023-01772-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/03/2023] [Indexed: 04/15/2023] Open
Abstract
Immunotherapy has emerged to play a rapidly expanding role in the treatment of cancers. Currently, many clinical trials of therapeutic agents are on ongoing with majority of immune checkpoint inhibitors (ICIs) especially programmed death receptor 1 (PD-1) and its ligand 1 (PD-L1) inhibitors. PD-1 and PD-L1, two main immune checkpoints, are expressed at high levels in thymic epithelial tumors (TETs) and could be predictors of the progression and immunotherapeutic efficacy of TETs. However, despite inspiring efficacy reported in clinical trials and clinical practice, significantly higher incidence of immune-related adverse events (irAEs) than other tumors bring challenges to the administration of ICIs in TETs. To develop safe and effective immunotherapeutic patterns in TETs, understanding the clinical properties of patients, the cellular and molecular mechanisms of immunotherapy and irAEs occurrence are crucial. In this review, the progress of both basic and clinical research on immune checkpoints in TETs, the evidence of therapeutic efficacy and irAEs based on PD-1 /PD-L1 inhibitors in TETs treatment are discussed. Additionally, we highlighted the possible mechanisms underlying irAEs, prevention and management strategies, the insufficiency of current research and some worthy research insights. High PD-1/PD-L1 expression in TETs provides a rationale for ICI use. Completed clinical trials have shown an encouraging efficacy of ICIs, despite the high rate of irAEs. A deeper mechanism understanding at molecular level how ICIs function in TETs and why irAEs occur will help maximize the immunotherapeutic efficacy while minimizing irAEs risks in TET treatment to improve patient prognosis.
Collapse
Affiliation(s)
- Yong-Qiang Ao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, 200032, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Gao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, 200032, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuai Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, 200032, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jia-Hao Jiang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, 200032, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jie Deng
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai, China
| | - Hai-Kun Wang
- CAS Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Bei Xu
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Jian-Yong Ding
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, 200032, Shanghai, China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
11
|
Yang CA, Li JP, Lai YH, Huang YL, Lin CY, Lan JL. Assessing the Immune Cell Subset and Genetic Mutations in Patients With Palindromic Rheumatism Seronegative for Rheumatoid Factor and Anti-Cyclic Citrullinated Peptide. Arthritis Rheumatol 2023; 75:187-200. [PMID: 35819819 DOI: 10.1002/art.42297] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 06/17/2022] [Accepted: 07/07/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The etiology underlying cases of palindromic rheumatism (PR) not associated with other rheumatic diseases in patients who are seronegative for rheumatoid factor and anti-cyclic citrullinated peptide (seronegative PR) is unclear. We aimed to investigate the immune cells and genes involved. METHODS This was a single-center comparative study of 48 patients with seronegative PR and 48 healthy controls. Mass cytometry and RNA sequencing were used to identify distinct immune cell subsets in blood. Among the 48 seronegative PR patients, plasma samples from 40 patients were evaluated by enzyme-linked immunosorbent assay for cytokine levels, and peripheral blood samples from 25 patients were evaluated by flow cytometry for mononuclear cell subsets. Plasma samples from 21 patients were evaluated by real-time polymerase chain reaction for differential gene and protein expression, and samples from 3 patients were analyzed with whole-exome sequencing for gene mutations. RESULTS Immunophenotyping revealed a markedly increased frequency of CD14+CD11b+CD36+ and CD4+CD25-CD69+ cells in seronegative PR patients with active flares compared with healthy controls (P < 0.0001 for both cell subset comparisons). Gene enrichment analyses of RNA-sequencing data from sorted CD14+CD11b+CD36+ and CD4+CD25-CD69+ cells showed involvement of the inflammatory/stress response, phagocytosis, and regulation of apoptosis functional pathways. Up-regulated expression of CXCL16 and IL10RA was observed in monocytes from PR patients. Up-regulation of PFKFB3, DDIT4, and TGFB1, and down-regulation of PDIA6 were found in monocytes and lymphocytes from PR patients with active flares and PR patients in intercritical periods. Plasma levels of S100A8/A9 and interleukin-1β were elevated in PR patients. Whole-exome sequencing revealed novel polygenic mutations in HACL1, KDM5A, RASAL1, HAVCR2, PRDM9, MBOAT4, and JRKL. CONCLUSION In seronegative PR patients, we identified a distinct CD14+CD11b+CD36+ cell subset that can induce an inflammatory response under stress and exert antiinflammatory effects after phagocytosis of apoptotic cells, and a CD4+CD25-CD69+ T cell subset with pro- and antiinflammatory properties. Individuals with genetic mutations involving epigenetic modification, potentiation and resolution of stress-induced inflammation/apoptosis, and a dysregulated endoplasmic reticulum stress response could be predisposed to seronegative PR.
Collapse
Affiliation(s)
- Chin-An Yang
- College of Medicine, China Medical University, Division of Laboratory Medicine, China Medical University Hsinchu Hospital, and Departments of Medical Education and Pediatrics, China Medical University Hsinchu Hospital, Zhubei City, Taiwan, and Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Taiwan
| | - Ju-Pi Li
- Rheumatic Diseases Research Center, China Medical University Hospital, Department of Pathology, School of Medicine, Chung Shan Medical University and Chung Shan Medical University Hospital, Taiwan
| | - Yi-Hua Lai
- College of Medicine, China Medical University, Rheumatic Diseases Research Center, China Medical University Hospital, and Rheumatology and Immunology Center, China Medical University Hospital, Taiwan
| | - Ya-Ling Huang
- Division of Laboratory Medicine, China Medical University Hsinchu Hospital, Zhubei City, Taiwan
| | - Chien-Yu Lin
- Division of Laboratory Medicine, China Medical University Hsinchu Hospital, Zhubei City, Taiwan
| | - Joung-Liang Lan
- College of Medicine, China Medical University, Rheumatic Diseases Research Center, China Medical University Hospital, and Rheumatology and Immunology Center, China Medical University Hospital, Taiwan
| |
Collapse
|
12
|
Kanannejad Z, Soleimanian S, Ghahramani Z, Sepahi N, Mohkam M, Alyasin S, Kheshtchin N. Immune checkpoint molecules in prevention and development of asthma. Front Immunol 2023; 14:1070779. [PMID: 36865540 PMCID: PMC9972681 DOI: 10.3389/fimmu.2023.1070779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
Allergic asthma is a respiratory disease initiated by type-2 immune responses characterized by secretion of alarmins, interleukin-4 (IL-4), IL-5, and IL-13, eosinophilic inflammation, and airway hyperresponsiveness (AHR). Immune checkpoints (ICPs) are inhibitory or stimulatory molecules expressed on different immune cells, tumor cells, or other cell types that regulate immune system activation and maintain immune homeostasis. Compelling evidence indicates a key role for ICPs in both the progression and prevention of asthma. There is also evidence of asthma development or exacerbation in some cancer patients receiving ICP therapy. The aim of this review is to provide an updated overview of ICPs and their roles in asthma pathogenesis, and to assess their implications as therapeutic targets in asthma.
Collapse
Affiliation(s)
- Zahra Kanannejad
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeede Soleimanian
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Ghahramani
- Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Najmeh Sepahi
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Milad Mohkam
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soheila Alyasin
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nasim Kheshtchin
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
13
|
Wang Z, Chen C, Su Y, Ke N. Function and characteristics of TIM‑4 in immune regulation and disease (Review). Int J Mol Med 2022; 51:10. [PMID: 36524355 PMCID: PMC9848438 DOI: 10.3892/ijmm.2022.5213] [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: 09/12/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022] Open
Abstract
T‑cell/transmembrane immunoglobulin and mucin domain containing 4 (TIM‑4) is a phosphatidylserine receptor that is mainly expressed on antigen‑presenting cells and is involved in the recognition and efferocytosis of apoptotic cells. TIM‑4 has been found to be expressed in immune cells such as natural killer T, B and mast cells and to participate in multiple aspects of immune regulation, suggesting that TIM‑4 may be involved in a variety of immune‑related diseases. Recent studies have confirmed that TIM‑4 is also abnormally expressed in a variety of malignant tumor cells and is closely associated with the occurrence and development of tumors and the tumor immune microenvironment. The present study aimed to describe the expression and functional characteristics of TIM‑4 in detail and to comprehensively discuss its role in pathophysiological processes such as infection, allergy, metabolism, autoimmunity and tumor immunity. The current review provided a comprehensive understanding of the functions and characteristics of TIM‑4, as well as novel ideas for the diagnosis and treatment of diseases.
Collapse
Affiliation(s)
- Ziyao Wang
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Chen Chen
- Department of Radiology, The First People's Hospital of Chengdu, Chengdu, Sichuan 610095, P.R. China
| | - Yingzhen Su
- Kunming University School of Medicine, Kunming University School, Kunming, Yunnan 650124, P.R. China
| | - Nengwen Ke
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China,Correspondence to: Professor Nengwen Ke, Department of Pancreatic Surgery, West China Hospital, Sichuan University, 37 Guoxue Lane, Chengdu, Sichuan 610041, P.R. China, E-mail:
| |
Collapse
|
14
|
Hattori T, Saito T, Miyamoto H, Kajihara M, Igarashi M, Takada A. Single Nucleotide Variants of the Human TIM-1 IgV Domain with Reduced Ability to Promote Viral Entry into Cells. Viruses 2022; 14:v14102124. [PMID: 36298679 PMCID: PMC9611583 DOI: 10.3390/v14102124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/18/2022] [Accepted: 09/21/2022] [Indexed: 11/29/2022] Open
Abstract
Human T-cell immunoglobulin mucin 1 (hTIM-1) is known to promote cellular entry of enveloped viruses. Previous studies suggested that the polymorphisms of hTIM-1 affected its function. Here, we analyzed single nucleotide variants (SNVs) of hTIM-1 to determine their ability to promote cellular entry of viruses using pseudotyped vesicular stomatitis Indiana virus (VSIV). We obtained hTIM-1 sequences from a public database (Ensembl genome browser) and identified 35 missense SNVs in 3 loops of the hTIM-1 immunoglobulin variable (IgV) domain, which had been reported to interact with the Ebola virus glycoprotein (GP) and phosphatidylserine (PS) in the viral envelope. HEK293T cells transiently expressing wildtype hTIM-1 or its SNV mutants were infected with VSIVs pseudotyped with filovirus or arenavirus GPs, and their infectivities were compared. Eleven of the thirty-five SNV substitutions reduced the efficiency of hTIM-1-mediated entry of pseudotyped VSIVs. These SNV substitutions were found not only around the PS-binding pocket but also in other regions of the molecule. Taken together, our findings suggest that some SNVs of the hTIM-1 IgV domain have impaired ability to interact with PS and/or viral GPs in the viral envelope, which may affect the hTIM-1 function to promote viral entry into cells.
Collapse
Affiliation(s)
- Takanari Hattori
- Division of Global Epidemiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan
| | - Takeshi Saito
- Division of Global Epidemiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan
| | - Hiroko Miyamoto
- Division of Global Epidemiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan
| | - Masahiro Kajihara
- Division of Global Epidemiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan
| | - Manabu Igarashi
- Division of Global Epidemiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan
| | - Ayato Takada
- Division of Global Epidemiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan
- Department of Disease Control, School of Veterinary Medicine, University of Zambia, Lusaka 10101, Zambia
- One Health Research Center, Hokkaido University, Sapporo 001-0020, Japan
- Correspondence: ; Tel.: +81-11-706-9502; Fax: +81-11-706-7310
| |
Collapse
|
15
|
Chen L, Qing J, Xiao Y, Huang X, Chi Y, Chen Z. TIM-1 promotes proliferation and metastasis, and inhibits apoptosis, in cervical cancer through the PI3K/AKT/p53 pathway. BMC Cancer 2022; 22:370. [PMID: 35392845 PMCID: PMC8991826 DOI: 10.1186/s12885-022-09386-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 03/07/2022] [Indexed: 12/09/2022] Open
Abstract
Background T-cell immunoglobulin mucin-1 (TIM-1) has been reported to be associated with the biological behavior of several malignant tumors; however, it is not clear whether it has a role in cervical cancer (CC). Methods TIM-1 expression in cervical epithelial tumor tissues and cells was detected by immunohistochemistry or real-time quantitative-PCR and western blotting. CC cells from cell lines expressing low levels of TIM-1 were infected with lentiviral vectors encoding TIM-1. Changes in the malignant behavior of CC cells were assessed by CCK-8, wound healing, Transwell migration and invasion assays, and flow cytometry in vitro; while a xenograft tumor model was established to analyze the effects of TIM-1 on tumor growth in vivo. Changes in the levels of proteins related to the cell cycle, apoptosis, and Epithelial-mesenchymal transition (EMT) were determined by western blotting. Results TIM-1 expression was higher in CC tissues, than in high grade squamous intraepithelial lesion, low grade squamous intraepithelial lesion, or normal cervical tissues, and was also expressed in three CC cell lines. In HeLa and SiHa cells overexpressing TIM-1, proliferation, invasion, and migration increased, while whereas apoptosis was inhibited. Furthermore, TIM-1 downregulated the expression of p53, BAX, and E-cadherin, and increased cyclin D1, Bcl-2, Snail1, N-cadherin, vimentin, MMP-2, and VEGF. PI3K, p-AKT, and mTOR protein levels also increased, while total AKT protein levels remained unchanged. Conclusions Our study indicated that TIM-1 overexpression promoted cell migration and invasion, and inhibited cell apoptosis in CC through modulation of the PI3K/AKT/p53 and PI3K/AKT/mTOR signaling pathways, and may be a candidate diagnostic biomarker of this disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09386-7.
Collapse
Affiliation(s)
- Liuyan Chen
- Joint Inspection Center of Precision Medicine, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, Guangxi, People's Republic of China.,Department of Clinical Laboratory, the first affiliated hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, People's Republic of China
| | - Jilin Qing
- Center for Reproductive Medicine and Genetics, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, Guangxi, People's Republic of China
| | - Yangyang Xiao
- Department of Clinical Laboratory, Binzhou Medical University Hospital, Binzhou, Shandong, People's Republic of China
| | - Xiaomei Huang
- Joint Inspection Center of Precision Medicine, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, Guangxi, People's Republic of China.,Guangxi University of Chinese Medicine, Nanning, Guangxi, People's Republic of China
| | - Yanlin Chi
- Department of Clinical Laboratory, the first affiliated hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, People's Republic of China
| | - Zhizhong Chen
- Joint Inspection Center of Precision Medicine, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, Guangxi, People's Republic of China.
| |
Collapse
|
16
|
Boehne C, Behrendt AK, Meyer-Bahlburg A, Boettcher M, Drube S, Kamradt T, Hansen G. Tim-3 is dispensable for allergic inflammation and respiratory tolerance in experimental asthma. PLoS One 2021; 16:e0249605. [PMID: 33822811 PMCID: PMC8023500 DOI: 10.1371/journal.pone.0249605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/19/2021] [Indexed: 11/18/2022] Open
Abstract
T cell immunoglobulin and mucin domain-containing molecule-3 (Tim-3) has been described as a transmembrane protein, expressed on the surface of various T cells as well as different cells of innate immunity. It has since been associated with Th1 mediated autoimmune diseases and transplantation tolerance studies, thereby indicating a possible role of this receptor in counter-regulation of Th2 immune responses. In the present study we therefore directly examined the role of Tim-3 in allergic inflammation and respiratory tolerance. First, Tim-3-/- mice and wild type controls were immunized and challenged with the model allergen ovalbumin (OVA) to induce an asthma-like phenotype. Analysis of cell numbers and distribution in the bronchoalveolar lavage (BAL) fluid as well as lung histology in H&E stained lung sections demonstrated a comparable degree of eosinophilic inflammation in both mouse strains. Th2 cytokine production in restimulated cell culture supernatants and serum IgE and IgG levels were equally increased in both genotypes. In addition, cell proliferation and the distribution of different T cell subsets were comparable. Moreover, analysis of both mouse strains in our respiratory tolerance model, where mucosal application of the model allergen before immunization, prevents the development of an asthma-like phenotype, revealed no differences in any of the parameters mentioned above. The current study demonstrates that Tim-3 is dispensable not only for the development of allergic inflammation but also for induction of respiratory tolerance in mice in an OVA-based model.
Collapse
Affiliation(s)
- Carolin Boehne
- Department of Pediatrics and Adolescent Medicine, Pediatric Pulmonology, Allergology and Neonatology, Hannover Medical School, Lower Saxony, Germany
| | - Ann-Kathrin Behrendt
- Department of Pediatrics and Adolescent Medicine, Pediatric Pulmonology, Allergology and Neonatology, Hannover Medical School, Lower Saxony, Germany
| | - Almut Meyer-Bahlburg
- Department of Pediatrics and Adolescent Medicine, Pediatric Pulmonology, Allergology and Neonatology, Hannover Medical School, Lower Saxony, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Lower Saxony, Germany
| | - Martin Boettcher
- Institute of Immunology, University Hospital Jena, Jena, Thuringia, Germany
| | - Sebastian Drube
- Institute of Immunology, University Hospital Jena, Jena, Thuringia, Germany
| | - Thomas Kamradt
- Institute of Immunology, University Hospital Jena, Jena, Thuringia, Germany
| | - Gesine Hansen
- Department of Pediatrics and Adolescent Medicine, Pediatric Pulmonology, Allergology and Neonatology, Hannover Medical School, Lower Saxony, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Lower Saxony, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Lower Saxony, Germany
- * E-mail:
| |
Collapse
|
17
|
Wagner M, Jasek M, Karabon L. Immune Checkpoint Molecules-Inherited Variations as Markers for Cancer Risk. Front Immunol 2021; 11:606721. [PMID: 33519815 PMCID: PMC7840570 DOI: 10.3389/fimmu.2020.606721] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/25/2020] [Indexed: 12/13/2022] Open
Abstract
In recent years, immunotherapy has been revolutionized by a new approach that works by blocking receptors called immune checkpoints (IC). These molecules play a key role in maintaining immune homeostasis, mainly by suppressing the immune response and by preventing its overactivation. Since inhibition of the immune response by IC can be used by cancer to avoid recognition and destruction by immune system, blocking them enhances the anti-tumor response. This therapeutic approach has brought spectacular clinical effects. The ICs present heterogeneous expression patterns on immune cells, which may affect the effectiveness of immunotherapy. The inherited genetic variants in regulatory regions of ICs genes can be considered as potential factors responsible for observed inter-individual differences in ICs expression levels on immune cells. Additionally, polymorphism located in exons may introduce changes to ICs amino acid sequences with potential impact on functional properties of these molecules. Since genetic variants may affect both expression and structure of ICs, they are considered as risk factors of cancer development. Inherited genetic markers such as SNPs may also be useful in stratification patients into groups which will benefit from particular immunotherapy. In this review, we have comprehensively summarized the current understanding of the relationship between inherited variations of CTLA-4, PDCD1, PD-L1, BTLA, TIM-3, and LAG-3 genes in order to select SNPs which can be used as predictive biomarkers in personalized evaluation of cancer risk development and outcomes as well as possible response to immunotherapy.
Collapse
Affiliation(s)
| | - Monika Jasek
- Laboratory of Genetics and Epigenetics of Human Diseases, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | | |
Collapse
|
18
|
Ding QQ, Chauvin JM, Zarour HM. Targeting novel inhibitory receptors in cancer immunotherapy. Semin Immunol 2020; 49:101436. [PMID: 33288379 DOI: 10.1016/j.smim.2020.101436] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/24/2022]
Abstract
T cells play a critical role in promoting tumor regression in both experimental models and humans. Yet, T cells that are chronically exposed to tumor antigen during cancer progression can become dysfunctional/exhausted and fail to induce tumor destruction. Such tumor-induced T cell dysfunction may occur via multiple mechanisms. In particular, immune checkpoint inhibitory receptors that are upregulated by tumor-infiltrating lymphocytes in many cancers limit T cell survival and function. Overcoming this inhibitory receptor-mediated T cell dysfunction has been a central focus of recent developments in cancer immunotherapy. Immunotherapies targeting inhibitory receptor pathways such as programmed cell death 1 (PD-1)/programmed death ligand 1 and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), alone or in combination, confer significant clinical benefits in multiple tumor types. However, many patients with cancer do not respond to immune checkpoint blockade, and dual PD-1/CTLA-4 blockade may cause serious adverse events, which limits its indications. Targeting novel non-redundant inhibitory receptor pathways contributing to tumor-induced T cell dysfunction in the tumor microenvironment may prove efficacious and non-toxic. This review presents preclinical and clinical findings supporting the roles of two key pathways-T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) and T cell immunoreceptor with Ig and ITIM domain (TIGIT)/CD226/CD96/CD112R-in cancer immunotherapy.
Collapse
Affiliation(s)
- Quan-Quan Ding
- Department of Medicine and Division of Hematology/Oncology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Joe-Marc Chauvin
- Department of Medicine and Division of Hematology/Oncology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Hassane M Zarour
- Department of Medicine and Division of Hematology/Oncology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
19
|
TIM-3 Genetic Variants Are Associated with Altered Clinical Outcome and Susceptibility to Gram-Positive Infections in Patients with Sepsis. Int J Mol Sci 2020; 21:ijms21218318. [PMID: 33171904 PMCID: PMC7664272 DOI: 10.3390/ijms21218318] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 11/04/2020] [Indexed: 12/29/2022] Open
Abstract
Background: Previous studies have reported the fundamental role of immunoregulatory proteins in the clinical phenotype and outcome of sepsis. This study investigated two functional single nucleotide polymorphisms (SNPs) of T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3), which has a negative stimulatory function in the T cell immune response. Methods: Patients with sepsis (n = 712) were prospectively enrolled from three intensive care units (ICUs) at the University Medical Center Goettingen since 2012. All patients were genotyped for the TIM-3 SNPs rs1036199 and rs10515746. The primary outcome was 28-day mortality. Disease severity and microbiological findings were secondary endpoints. Results: Kaplan-Meier survival analysis demonstrated a significantly lower 28-day mortality for TIM-3 rs1036199 AA homozygous patients compared to C-allele carriers (18% vs. 27%, p = 0.0099) and TIM-3 rs10515746 CC homozygous patients compared to A-allele carriers (18% vs. 26%, p = 0.0202). The TIM-3 rs1036199 AA genotype and rs10515746 CC genotype remained significant predictors for 28-day mortality in the multivariate Cox regression analysis after adjustment for relevant confounders (adjusted hazard ratios: 0.67 and 0.70). Additionally, patients carrying the rs1036199 AA genotype presented more Gram-positive and Staphylococcus epidermidis infections, and rs10515746 CC homozygotes presented more Staphylococcus epidermidis infections. Conclusion: The studied TIM-3 genetic variants are associated with altered 28-day mortality and susceptibility to Gram-positive infections in sepsis.
Collapse
|
20
|
Khan S, Gerber DE. Autoimmunity, checkpoint inhibitor therapy and immune-related adverse events: A review. Semin Cancer Biol 2020; 64:93-101. [PMID: 31330185 PMCID: PMC6980444 DOI: 10.1016/j.semcancer.2019.06.012] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 06/25/2019] [Indexed: 02/08/2023]
Abstract
Immune checkpoint inhibitors have emerged as a remarkable treatment option for diverse cancer types. However, a significant number of patients on checkpoint inhibitors develop immune-related adverse events (irAEs) affecting a wide variety of organs. These events, which may reflect enhanced T cell activation, are unpredictable, heterogeneous, and in some instances permanent or life-threatening. It is not clear whether these toxicities are distinct from conventional autoimmune diseases or whether the manifestation of irAEs is associated with therapeutic efficacy. Studies across the spectrum of basic, preclinical and clinical research deciphering the role of genetics, epigenetics, gut microbiota and underlying immune status of patients who develop irAEs are required to gain a deeper mechanistic understanding. Insights gained from such studies will facilitate identification of biomarkers for optimal treatment and clinical management of patients. In this Review, we provide basic and clinical understanding of immune checkpoint inhibitors and irAEs. We discuss the connection between immune system, autoimmunity and cancer; immune checkpoint inhibitors and associated autoimmune toxicities; insights into potential underlying mechanisms of irAEs; impact of autoimmune diagnosis on cancer outcome; and management of irAEs.
Collapse
Affiliation(s)
- Shaheen Khan
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, 75390-9093, United States.
| | - David E Gerber
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390-9093, United States; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390-9093, United States; Department of Population & Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390-9093, United States.
| |
Collapse
|
21
|
Wu JL, Zhao J, Zhang HB, Zuo WW, Li Y, Kang S. Genetic variants and expression of the TIM-3 gene are associated with clinical prognosis in patients with epithelial ovarian cancer. Gynecol Oncol 2020; 159:270-276. [PMID: 32694063 DOI: 10.1016/j.ygyno.2020.07.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/06/2020] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Polymorphisms of T cell immunoglobulin and mucin domain-containing molecule 3 (TIM-3) were reported to be associated with cancer risk and patients' survival. This study aims to investigate the correlation of TIM-3 polymorphisms with susceptibility to epithelial ovarian cancer (EOC) and patients' outcomes. METHODS A total of 700 EOC patients and 710 healthy controls from North China were included. The polymorphisms (rs10053538, rs10515746 and rs1036199) were genotyped using the polymerase chain reaction/ligase detection reaction (PCR-LDR) method. Survival data were available for 339 patients after cytoreductive surgery. The expression level of TIM-3 was detected by real-time quantitative PCR (RT-qPCR). The prognostic value of TIM3 in EOC patients was assessed using the Kaplan-Meier plotter database. RESULTS The results showed that none of the TIM3 polymorphisms were associated with the risk of developing EOC. Patients with the rs10053538 CA + AA genotype had worse PFS and OS than those with the CC genotype (HR = 1.49, 95% CI = 1.05-2.09, P = 0.024 and HR = 1.57, 95%CI = 1.09-2.26, P = 0.017, respectively). The RT-qPCR results showed that the expression levels of TIM-3 mRNA in EOC tissues with the rs10053538CA + AA genotypes were significantly higher than those with the CC genotype (P = 0.006). Analysis using the Kaplan-Meier plotter database showed that high expression of TIM-3 mRNA was significantly associated with shorter PFS and OS in EOC patients (HR = 1.57, 95%CI = 1.29-1.91, P < 0.001 and HR = 1.31, 95% CI = 1.06-1.63, P = 0.013, respectively). CONCLUSIONS TIM-3 polymorphisms were not associated with risk of developing EOC. Both rs10053538 and the expression level of TIM-3 mRNA may be associated with its clinical outcome in EOC patients.
Collapse
Affiliation(s)
- Jian-Lei Wu
- Department of Obstetrics and Gynaecology, Hebei Medical University, Fourth Hospital, Shijiazhuang, China
| | - Jian Zhao
- Department of Obstetrics and Gynaecology, First Hospital of Shijiazhuang, Shijiazhuang, China
| | - Hai-Bo Zhang
- Department of Obstetrics and Gynaecology, Hebei Medical University, Fourth Hospital, Shijiazhuang, China
| | - Wei-Wei Zuo
- Department of Obstetrics and Gynaecology, Hebei Medical University, Fourth Hospital, Shijiazhuang, China
| | - Yan Li
- Department of Molecular Biology, Hebei Medical University, Fourth Hospital, Shijiazhuang, China.
| | - Shan Kang
- Department of Obstetrics and Gynaecology, Hebei Medical University, Fourth Hospital, Shijiazhuang, China.
| |
Collapse
|
22
|
Li J, Qiu Y, Li L, Wang J, Cheuk YC, Sang R, Jia Y, Wang J, Zhang Y, Rong R. Histone Methylation Inhibitor DZNep Ameliorated the Renal Ischemia-Reperfusion Injury via Inhibiting TIM-1 Mediated T Cell Activation. Front Med (Lausanne) 2020; 7:305. [PMID: 32754604 PMCID: PMC7365856 DOI: 10.3389/fmed.2020.00305] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 05/27/2020] [Indexed: 12/17/2022] Open
Abstract
Renal ischemia-reperfusion injury (IRI) after renal transplantation often leads to the loss of kidney graft function. However, there is still a lack of efficient regimens to prevent or alleviate renal IRI. Our study focused on the renoprotective effect of 3-Deazaneplanocin A (DZNep), which is a histone methylation inhibitor. We found that DZNep significantly alleviated renal IRI by suppressing nuclear factor kappa-B (NF-κB), thus inhibiting the expression of inflammatory factors in renal tubular epithelial cells in vivo or in vitro. After treatment with DZNep, T cell activation was impaired in the spleen and kidney, which correlated with the downregulated expression of T-cell immunoglobulin mucin (TIM)-1 on T cells and TIM-4 in macrophages. In addition, pretreatment with DZNep was not sufficient to protect the kidney, while administration of DZNep from before to after surgery significantly ameliorated IRI. Our findings suggest that DZNep can be a novel strategy for preventing renal IRI following kidney transplantation.
Collapse
Affiliation(s)
- Jiawei Li
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Yue Qiu
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, China.,Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Long Li
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiyan Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Yin Celeste Cheuk
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Ruirui Sang
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Yichen Jia
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Jina Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Yi Zhang
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, China.,Biomedical Research Center, Institute for Clinical Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ruiming Rong
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| |
Collapse
|
23
|
Acharya N, Sabatos-Peyton C, Anderson AC. Tim-3 finds its place in the cancer immunotherapy landscape. J Immunother Cancer 2020; 8:e000911. [PMID: 32601081 PMCID: PMC7326247 DOI: 10.1136/jitc-2020-000911] [Citation(s) in RCA: 228] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2020] [Indexed: 02/06/2023] Open
Abstract
The blockade of immune checkpoint receptors has made great strides in the treatment of major cancers, including melanoma, Hodgkin's lymphoma, renal, and lung cancer. However, the success rate of immune checkpoint blockade is still low and some cancers, such as microsatellite-stable colorectal cancer, remain refractory to these treatments. This has prompted investigation into additional checkpoint receptors. T-cell immunoglobulin and mucin domain 3 (Tim-3) is a checkpoint receptor expressed by a wide variety of immune cells as well as leukemic stem cells. Coblockade of Tim-3 and PD-1 can result in reduced tumor progression in preclinical models and can improve antitumor T-cell responses in cancer patients. In this review, we will discuss the basic biology of Tim-3, its role in the tumor microenvironment, and the emerging clinical trial data that point to its future application in the field of immune-oncology.
Collapse
Affiliation(s)
- Nandini Acharya
- Department of Neurology, Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Catherine Sabatos-Peyton
- Exploratory Immuno-oncology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USA
| | - Ana Carrizosa Anderson
- Department of Neurology, Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
24
|
Shen H, Zhang X, Al Hafiz MA, Liang X, Yao Q, Guo M, Xu G, Zhong X, Zhou Q, Zhao H. The Proteins Interacting with Prmt5 in Medaka (Oryzias latipes) Identified by Yeast Two-Hybridization. Protein Pept Lett 2020; 27:971-978. [PMID: 32370700 DOI: 10.2174/0929866527666200505213431] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/08/2020] [Accepted: 03/31/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Prmt5 plays major role in regulation of gene expression, RNA processing, cell growth and differentiation, signal transduction, germ cell development, etc., in mammals. Prmt5 is also related to cancer. Knowing the proteins interacting with Prmt5 is important to understand Prmt5's function in cells. Although there have been reports on proteins binding with Prmt5 in mammals, the partner proteins of Prmt5 in fish are still unclear. OBJECTIVES The objective was to obtain proteins that bind with Prmt5 in medaka, a model fish. METHODS Yeast two hybridization was adopted to achieve the objective. Medaka Prmt5 was used as a bait to fish the prey, binding proteins in a cDNA library of medaka. Co-immunoprecipitation and in silicon analysis were performed to study the interaction of medaka Mep50 and Prmt5. RESULTS Eight proteins were identified to bind with Prmt5 from 69 preliminary positive colonies. The binding proteins are methylosome protein 50 (Mep50), apolipoprotein A-I-like (Apo-AI), PR domain containing protein 1a with zinc fingers (Prdm1a), Prdm1b, T-cell immunoglobulin mucin family member 3 (Tim-3), phosphoribosylaminoimidazole carboxylase and phosphoribosylaminoimidazolesuccinocarboxamide synthase (Paics), NADH dehydrogenase subunit 4 (ND4) and sciellin (Scl). Co-immunoprecipitation confirmed the interaction of medaka Prmt5 and Mep50. Predicted structures of medaka Prtm5 and Mep50 are similar to that of human PRMT5 and MEP50. CONCLUSION Medaka Mep50, Prdm1a, Prdm1b, Apo-AI, Tim-3, Paics, ND4, and Scl bind with Prmt5.
Collapse
Affiliation(s)
- Hao Shen
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Xiaosha Zhang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Md Abdullah Al Hafiz
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Xiaoting Liang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Qiting Yao
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Maomao Guo
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Gongyu Xu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Xueping Zhong
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Qingchun Zhou
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Haobin Zhao
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| |
Collapse
|
25
|
Schnell A, Bod L, Madi A, Kuchroo VK. The yin and yang of co-inhibitory receptors: toward anti-tumor immunity without autoimmunity. Cell Res 2020; 30:285-299. [PMID: 31974523 PMCID: PMC7118128 DOI: 10.1038/s41422-020-0277-x] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 01/06/2020] [Indexed: 12/31/2022] Open
Abstract
Co-inhibitory receptors are important regulators of T-cell function that define the balance between tolerance and autoimmunity. The immune regulatory function of co-inhibitory receptors, including CTLA-4, PD-1, TIM-3, TIGIT, and LAG-3, was first discovered in the setting of autoimmune disease models, in which their blockade or deficiency resulted in induction or exacerbation of the disease. Later on, co-inhibitory receptors on lymphocytes have also been found to influence outcomes in tumor and chronic viral infection settings. These receptors suppress T-cell function in the tumor microenvironment (TME), thereby making the T cells dysfunctional. Based on this observation, blockade of co-inhibitory receptors (also known as checkpoint molecules) has emerged as a successful treatment option for a number of human cancers. However, severe autoimmune-like side effects limit the use of therapeutics that block individual or combinations of co-inhibitory receptors for cancer treatment. In this review we provide an overview of the role of co-inhibitory receptors in autoimmunity and anti-tumor immunity. We then discuss current approaches and future directions to leverage our knowledge of co-inhibitory receptors to target them in tumor immunity without inducing autoimmunity.
Collapse
Affiliation(s)
- Alexandra Schnell
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Lloyd Bod
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Asaf Madi
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| |
Collapse
|
26
|
Abstract
T cell immunoglobulin and mucin domain-containing protein 3 (TIM3), a member of the TIM family, was originally identified as a receptor expressed on interferon-γ-producing CD4+ and CD8+ T cells. Initial data indicated that TIM3 functioned as a 'co-inhibitory' or 'checkpoint' receptor, but due to the lack of a definable inhibitory signalling motif, it was also suggested that TIM3 might act as a co-stimulatory receptor. Recent studies have shown that TIM3 is part of a module that contains multiple co-inhibitory receptors (checkpoint receptors), which are co-expressed and co-regulated on dysfunctional or 'exhausted' T cells in chronic viral infections and cancer. Furthermore, co-blockade of TIM3 and programmed cell death 1 (PD1) can result in tumour regression in preclinical models and can improve anticancer T cell responses in patients with advanced cancers. Here, we highlight the developments in understanding TIM3 biology, including novel ligand identification and the discovery of loss-of-function mutations associated with human disease. In addition, we summarize emerging data from human clinical trials showing that TIM3 indeed acts as a 'checkpoint' receptor and that inhibition of TIM3 enhances the antitumour effect of PD1 blockade.
Collapse
|
27
|
Ataei M, Behfarjam F, Jadali Z. TIM-3 genetic variants and risk of Behçet disease in the Iranian population. An Bras Dermatol 2019; 94:429-433. [PMID: 31644615 PMCID: PMC7007023 DOI: 10.1590/abd1806-4841.20198022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Accepted: 11/08/2018] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Behçet disease is a prototypical systemic autoimmune disease, caused by a complex interplay between environmental and genetic factors. The transmembrane immunoglobulin and mucin domain-3 (TIM-3) is a distinct member of the TIM family that is preferentially expressed on Th1 cells and plays a role in Th1-mediated autoimmune or inflammatory diseases, such as Behçet disease. OBJECTIVE The aim of this study was to test the potential association between TIM-3 gene polymorphisms and Behçet disease. METHODS Two single-nucleotide polymorphisms of TIM-3 (rs9313439 and rs10515746) were genotyped in 212 patients with Behçet disease and 200 healthy controls. Typing of the polymorphisms was performed using multiplex PCR amplification. RESULTS There were no significant differences in allele and genotype frequencies between the Behçet disease patients and controls who were successfully genotyped. Similar results were also found after stratification by gender, age, or clinical features. STUDY LIMITATIONS Lack of studies on various racial or ethnic groups and small sample size. CONCLUSION This study failed to demonstrate any association between the tested TIM-3 polymorphisms and Behçet disease.
Collapse
Affiliation(s)
- Mitra Ataei
- Clinical Genetics Department, National Institute of Genetic Engineering and Biotechnology Tehran, Iran
| | - Farinaz Behfarjam
- Clinical Genetics Department, National Institute of Genetic Engineering and Biotechnology Tehran, Iran
| | - Zohreh Jadali
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Fang H, Yuan C, Gu X, Chen Q, Huang D, Li H, Sun M. Association between TIM-3 polymorphisms and cancer risk: a meta-analysis. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:550. [PMID: 32133384 PMCID: PMC6861791 DOI: 10.21037/atm.2019.09.101] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 09/10/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Single nucleotide polymorphisms (SNPs) of T-cell immunoglobulin- and mucin-domain-containing molecule 3 (TIM-3) were reported to individually associate with cancer risk. To further verify its correlation with human cancers, we evaluated the association of TIM-3 polymorphisms and the risk of cancer. METHODS Data were collected from electronic databases. Two reviewers independently selected studies, extracted data and assessed quality of the studies. Data were meta-analyzed using the STATA 13.0 software. Crude odd ratio (OR) and 95% confidence interval was used to estimate the association between TIM-3 polymorphism and cancer susceptibility. RESULTS All eligible case-control studies included a total of 4,852 participants (2,229 cases and 2,623 controls). The meta-analysis showed that TIM-3 SNPs (-1516G/T, -574G/T, +4259T/G, and haplotypes) were significantly associated with an increased risk of susceptibility toward all cancers. The subgroup analyses based on cancer types showed that TIM-3 -1516G/T SNP was only associated with an increased risk in developing cancers in the digestive system or in hospital-based populations. Moreover, the TIM-3 -574G/T SNP was associated with an increased cancer risk in the digestive system or other systems, while TIM-3 +4259T/G SNP was only associated with an increased cancer risk in hospital-based populations. Among the four haplotypes observed (GGT, TGT, GGG, and GTT), The GGG haplotype showed an increase in the odds of cancer by 2.614-fold (OR 2.614; 95% CI: 1.756-3.893) compared with the GGT haplotype. CONCLUSIONS TIM-3 SNPs (-1516G/T, -574G/T, +4259T/G and the four haplotypes) were associated with an increased risk of developing human cancers.
Collapse
Affiliation(s)
- Hongyan Fang
- Department of Oncology, The Fifth Hospital of Wuhan, Wuhan 430050, China
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Cheng Yuan
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Xinsheng Gu
- College of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Qiuju Chen
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
- Institute of Anesthesiology, Department of anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Dong Huang
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Heng Li
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Min Sun
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
- Institute of Anesthesiology, Department of anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| |
Collapse
|
29
|
Khan A, Bresnick A, Cahill S, Girvin M, Almo S, Quinn R. Advantages of Molecular Weight Identification during Native MS Screening. PLANTA MEDICA 2018; 84:1201-1212. [PMID: 29742762 PMCID: PMC6195442 DOI: 10.1055/a-0608-4870] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Native mass spectrometry detection of ligand-protein complexes allowed rapid detection of natural product binders of apo and calcium-bound S100A4 (a member of the metal binding protein S100 family), T cell/transmembrane, immunoglobulin (Ig), and mucin protein 3, and T cell immunoreceptor with Ig and ITIM (immunoreceptor tyrosine-based inhibitory motif) domains precursor protein from extracts and fractions. Based on molecular weight common hits were detected binding to all four proteins. Seven common hits were identified as apigenin 6-C-β-D-glucoside 8-C-α-L-arabinoside, sweroside, 4',5-dihydroxy-7-methoxyflavanone-6-C-rutinoside, loganin acid, 6-C-glucosylnaringenin, biochanin A 7-O-rutinoside and quercetin 3-O-rutinoside. Mass guided isolation and NMR identification of hits confirmed the mass accuracy of the ligand in the ligand-protein MS complexes. Thus, molecular weight ID from ligand-protein complexes by electrospray ionization Fourier transform mass spectrometry allowed rapid dereplication. Native mass spectrometry using electrospray ionization Fourier transform mass spectrometry is a tool for dereplication and metabolomics analysis.
Collapse
Affiliation(s)
- Ahad Khan
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, Australia
| | | | - Sean Cahill
- Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mark Girvin
- Albert Einstein College of Medicine, Bronx, NY, USA
| | - Steve Almo
- Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ronald Quinn
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, Australia
| |
Collapse
|
30
|
Pouladian M, Ganjalikhani-Hakemi M, Alsahebfosul F, Homayouni V, Khosravi S, Etemadifar M, Mazrouei F, Salehi R. The +4259A>C polymorphism of TIM-3 but not -1637C>T polymorphism of TIM-1 is associated with Multiple sclerosis in Isfahan population. Mult Scler Relat Disord 2017; 18:152-156. [PMID: 29141799 DOI: 10.1016/j.msard.2017.09.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 09/24/2017] [Accepted: 09/27/2017] [Indexed: 02/08/2023]
|
31
|
Homayouni V, Khanahmad H, Ganjalikhani-Hakemi M, Behdani M, Ghasemi P, Rezaei A. Stimulation of Camel Polyclonal Antibody against Human T cell Immunoglobulin and Mucin 3. IRANIAN JOURNAL OF BIOTECHNOLOGY 2017; 15:166-171. [PMID: 29845065 PMCID: PMC5811063 DOI: 10.15171/ijb.1427] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 05/14/2017] [Accepted: 09/11/2017] [Indexed: 02/05/2023]
Abstract
Background: T cell Immunoglobulin, Mucin (TIM)-3, is a type I transmembrane glycoprotein belonging to TIM family. This receptor expresses on T helper type 1 (Th1) cells that binds to galectin-9 (Gal9); inducing an inhibitory signal. As a result, apoptosis of Th1 cells occurs and cytotoxicity of CD8 T cells becomes evident in vitro. Therefore, this immunomodulatory molecule may be used as a novel target for clinical purposes. The production of camel polyclonal antibodies against TIM-3-expressing cell line was the purpose of this study. Objectives: In this study, we aimed to use HEK 293 cells expressing human TIM-3 to obtain camel polyclonal antibody against TIM-3 by immunization. Materials and Methods: A pre-synthesized human TIM-3cDNA was inserted into pcDNA3.1 plasmid and the new construct was transfected in HEK cell. TIM-3 expression was confirmed by qRT-PCR and flow cytometry. A camel (6 months old) was immunized with the lysate prepared from rTIM-3 expressing HEK cells 4 times. The anti-TIM-3 antibody level was evaluated using ELISA method. Results: TIM-3 was successfully cloned in HEK cells with 88% success rate. High level of anti-TIM-3 antibody was detected in the serum of the camel immunized with the recombinant cell lysate, after final injection. Conclusions: Our rhTIM-3 cell display system can be useful for future diagnostic or therapeutic approaches.
Collapse
Affiliation(s)
- Vida Homayouni
- Immunology Department, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Khanahmad
- Genetic Department, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Mahdi Behdani
- Biotechnology Research Center, Biotechnology Department, Venom & Biotherapeutics Molecules Lab, Pasteur Institute of Iran, Tehran, P.O.Box: 1316543551, Iran
| | - Pouria Ghasemi
- Immunology Department, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abbas Rezaei
- Immunology Department, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
32
|
Abstract
To limit excessive T cell-mediated inflammatory responses, the immune system has a milieu of inhibitory receptors, called immune checkpoints. Cancer cells have evolved to seize those inhibitory pathways and to prevent T cell-mediated killing of tumor cells. Therefore, immune checkpoint inhibitors (ICI) consisting of blocking antibodies against these receptors present an exciting avenue in the fight against cancer. The last decade has seen the implementation of ICI against a variety of cancer indications that have improved the overall anti-tumor responses and patient survival. However, inflammatory toxicities and autoimmunity are a significant adverse event of ICI therapies. In this review, we will discuss the biology of immune checkpoints, highlight research strategies that may help reduce the incidence of immune-related adverse events associated with ICI therapies, and also suggest investigational approaches to manipulate immune checkpoints to treat primary autoimmune disorders.
Collapse
Affiliation(s)
- Anna S Tocheva
- Department of Medicine, NYU School of Medicine, 450 E 29th Street, Room 806, New York, NY, 10016, USA. .,Perlmutter Cancer Center, NYU School of Medicine, New York, NY, 10016, USA.
| | - Adam Mor
- Department of Medicine, NYU School of Medicine, 450 E 29th Street, Room 806, New York, NY, 10016, USA. .,Perlmutter Cancer Center, NYU School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
33
|
Kalantari S, Nafar M. A comprehensive narrative review of diagnostic biomarkers in human primary membranous nephropathy. Biomark Med 2017; 11:781-797. [DOI: 10.2217/bmm-2017-0081] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Membranous nephropathy (MN) is relatively major cause of nephrotic syndrome in adults which is recognized as an organ-specific autoimmune disease. The etiology of most cases is idiopathic, whereas the secondary MN is caused by systemic autoimmune diseases, infections, medications and malignancies. The idiopathic disease is developed by the formation of sub-epithelial immune complex deposits most likely due to binding the circulating auto-antibodies to intrinsic antigen on podocytes. The major auto antibody is the anti-phospholipase A2 receptor (anti-PLA2R), however, it is not enough sensitive. Several attempts for diagnostic biomarker identification by modern analytical technologies have been devoted recently. This article reviews the biomarker candidates for primary type of MN that are detected by different approaches on human subjects.
Collapse
Affiliation(s)
- Shiva Kalantari
- Chronic Kidney Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Nafar
- Urology Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Thomas LJ, Vitale L, O'Neill T, Dolnick RY, Wallace PK, Minderman H, Gergel LE, Forsberg EM, Boyer JM, Storey JR, Pilsmaker CD, Hammond RA, Widger J, Sundarapandiyan K, Crocker A, Marsh HC, Keler T. Development of a Novel Antibody-Drug Conjugate for the Potential Treatment of Ovarian, Lung, and Renal Cell Carcinoma Expressing TIM-1. Mol Cancer Ther 2016; 15:2946-2954. [PMID: 27671527 DOI: 10.1158/1535-7163.mct-16-0393] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/15/2016] [Accepted: 09/18/2016] [Indexed: 11/16/2022]
Abstract
T-cell immunoglobulin and mucin domain 1 (TIM-1) is a type I transmembrane protein that was originally described as kidney injury molecule 1 (KIM-1) due to its elevated expression in kidney and urine after renal injury. TIM-1 expression is also upregulated in several human cancers, most notably in renal and ovarian carcinomas, but has very restricted expression in healthy tissues, thus representing a promising target for antibody-mediated therapy. To this end, we have developed a fully human monoclonal IgG1 antibody specific for the extracellular domain of TIM-1. This antibody was shown to bind purified recombinant chimeric TIM-1-Fc protein and TIM-1 expressed on a variety of transformed cell lines, including Caki-1 (human renal clear cell carcinoma), IGROV-1 (human ovarian adenocarcinoma), and A549 (human lung carcinoma). Internalization studies using confocal microscopy revealed the antibody was rapidly internalized by cells in vitro, and internalization was confirmed by quantitative imaging flow cytometry. An antibody-drug conjugate (ADC) was produced with the anti-TIM-1 antibody covalently linked to the potent cytotoxin, monomethyl auristatin E (MMAE), and designated CDX-014. The ADC was shown to exhibit in vitro cytostatic or cytotoxic activity against a variety of TIM-1-expressing cell lines, but not on TIM-1-negative cell lines. Using the Caki-1, IGROV-1, and A549 xenograft mouse models, CDX-014 showed significant antitumor activity in a clinically relevant dose range. Safety evaluation in nonhuman primates has demonstrated a good profile and led to the initiation of clinical studies of CDX-014 in renal cell carcinoma and potentially other TIM-1-expressing tumors. Mol Cancer Ther; 15(12); 2946-54. ©2016 AACR.
Collapse
Affiliation(s)
| | | | | | - Ree Y Dolnick
- Flow and Image Cytometry Facility, Roswell Park Cancer Institute, Buffalo, New York
| | - Paul K Wallace
- Flow and Image Cytometry Facility, Roswell Park Cancer Institute, Buffalo, New York
| | - Hans Minderman
- Flow and Image Cytometry Facility, Roswell Park Cancer Institute, Buffalo, New York
| | | | | | | | | | | | | | | | | | | | | | - Tibor Keler
- Celldex Therapeutics, Inc., Hampton, New Jersey
| |
Collapse
|
35
|
Mazrouei F, Ganjalikhani-Hakemi M, Salehi R, Alesahebfosoul F, Etemadifar M, Pouladian M, Meshkat R, Nekoueian S, Zarkesh-Esfahani H, Ziyaee-Ghahnaviyeh M. Association ofTIM-1 5383-5397ins/delandTIM-3 -1541C>T polymorphismswith multiple sclerosis in Isfahan population. Int J Immunogenet 2016; 43:131-4. [PMID: 27091308 DOI: 10.1111/iji.12264] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 03/06/2016] [Accepted: 04/04/2016] [Indexed: 02/05/2023]
Affiliation(s)
- F. Mazrouei
- Department of Immunology; School of Medicine; Isfahan University of Medical Science; Isfahan Iran
| | - M. Ganjalikhani-Hakemi
- Department of Immunology; School of Medicine; Isfahan University of Medical Science; Isfahan Iran
| | - R. Salehi
- Department of Cellular & Molecular Biology; School of Medicine; Isfahan University of Medical Science; Isfahan Iran
| | - F. Alesahebfosoul
- Department of Immunology; School of Medicine; Isfahan University of Medical Science; Isfahan Iran
| | - M. Etemadifar
- Department of Neurosciences; Al-Zahra Hospital; Isfahan University of Medical Science; Isfahan Iran
| | - M. Pouladian
- Department of Immunology; School of Medicine; Isfahan University of Medical Science; Isfahan Iran
| | - R. Meshkat
- Department of Immunology; School of Medicine; Shahrekord University of Medical Science; Sharekord Iran
| | - Sh. Nekoueian
- Department of Molecular Diagnosis; Provincial Health Center; Isfahan University of Medical Science; Isfahan Iran
| | - H. Zarkesh-Esfahani
- Department of Biology; School of Sciences; University of Isfahan; Isfahan Iran
| | - M. Ziyaee-Ghahnaviyeh
- Department of Immunology; School of Medicine; Isfahan University of Medical Science; Isfahan Iran
| |
Collapse
|
36
|
Abstract
Polymorphisms in the T cell (or transmembrane) immunoglobulin and mucin
domain 1 ( TIM-1) gene, particularly in the mucin
domain, have been associated with atopy and allergic diseases in mice and human.
Genetic- and antibody-mediated studies revealed that Tim-1 functions as a
positive regulator of Th2 responses, while certain antibodies to Tim-1 can
exacerbate or reduce allergic lung inflammation. Tim-1 can also positively
regulate the function of B cells, NKT cells, dendritic cells and mast cells.
However, the precise molecular mechanisms by which Tim-1 modulates immune cell
function are currently unknown. In this study, we have focused on defining
Tim-1-mediated signaling pathways that enhance mast cell activation through the
high affinity IgE receptor (FceRI). Using a Tim-1 mouse model lacking the mucin
domain (Tim-1 Dmucin), we show for the first time that the
polymorphic Tim-1 mucin region is dispensable for normal mast cell activation.
We further show that Tim-4 cross-linking of Tim-1 enhances select signaling
pathways downstream of FceRI in mast cells, including mTOR-dependent signaling,
leading to increased cytokine production but without affecting
degranulation.
Collapse
Affiliation(s)
- Binh Phong
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, USA.,Immunology Graduate Program, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Lawrence P Kane
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, USA
| |
Collapse
|
37
|
Brooks CR, Yeung MY, Brooks YS, Chen H, Ichimura T, Henderson JM, Bonventre JV. KIM-1-/TIM-1-mediated phagocytosis links ATG5-/ULK1-dependent clearance of apoptotic cells to antigen presentation. EMBO J 2015; 34:2441-64. [PMID: 26282792 DOI: 10.15252/embj.201489838] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 07/01/2015] [Indexed: 12/14/2022] Open
Abstract
Phagocytosis of apoptotic cells by both professional and semi-professional phagocytes is required for resolution of organ damage and maintenance of immune tolerance. KIM-1/TIM-1 is a phosphatidylserine receptor that is expressed on epithelial cells and can transform the cells into phagocytes. Here, we demonstrate that KIM-1 phosphorylation and association with p85 results in encapsulation of phagosomes by lipidated LC3 in multi-membrane organelles. KIM-1-mediated phagocytosis is not associated with increased ROS production, and NOX inhibition does not block LC3 lipidation. Autophagy gene expression is required for efficient clearance of apoptotic cells and phagosome maturation. KIM-1-mediated phagocytosis leads to pro-tolerogenic antigen presentation, which suppresses CD4 T-cell proliferation and increases the percentage of regulatory T cells in an autophagy gene-dependent manner. Taken together, these data reveal a novel mechanism of epithelial biology linking phagocytosis, autophagy and antigen presentation to regulation of the inflammatory response.
Collapse
Affiliation(s)
- Craig R Brooks
- Department of Medicine, Renal Division, Brigham and Women's Hospital Harvard Medical School, Boston, MA, USA
| | - Melissa Y Yeung
- Department of Medicine, Renal Division, Brigham and Women's Hospital Harvard Medical School, Boston, MA, USA Transplantation Research Center, Brigham and Women's Hospital Harvard Medical School, Boston, MA, USA
| | - Yang S Brooks
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, MA, USA Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | - Hui Chen
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Takaharu Ichimura
- Department of Medicine, Renal Division, Brigham and Women's Hospital Harvard Medical School, Boston, MA, USA
| | - Joel M Henderson
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Joseph V Bonventre
- Department of Medicine, Renal Division, Brigham and Women's Hospital Harvard Medical School, Boston, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA
| |
Collapse
|
38
|
Wu M, Zhu Y, Zhao J, Ai H, Gong Q, Zhang J, Zhao J, Wang Q, La X, Ding J. Soluble costimulatory molecule sTim3 regulates the differentiation of Th1 and Th2 in patients with unexplained recurrent spontaneous abortion. Int J Clin Exp Med 2015; 8:8812-8819. [PMID: 26309533 PMCID: PMC4537953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 06/02/2015] [Indexed: 06/04/2023]
Abstract
This study is to investigate the mechanism of unexplained recurrent spontaneous abortion (URSA). A total of 35 cases of URSA patients (URSA group), 20 cases with normal pregnancy (normal pregnancy group) and 20 healthy non-pregnancy candidates (healthy control group) were enrolled in this study. Enzyme linked immunosorbent assay (ELISA) method was used for detection of serum soluble Tim-3 (sTim3) and Galectin-9. Cytokine bead array (CBA) determination method was used to detect IFN-γ and IL-4 expression levels. Compared with the healthy control group, sTim-3 levels in normal pregnancy group and URSA group increased, and URSA group had significantly higher sTim-3 levels than normal pregnancy group (P < 0.05). Compared with the healthy control group, Galectin-9 levels in normal pregnancy group and URSA group also increased. However, the normal pregnancy group had significantly higher Galectin-9 level than URSA group (P < 0.05). IFN-γ levels in normal pregnancy group and URSA group were lower than those in healthy control group, and IFN-γ levels in the normal pregnancy group were significantly lower than those in URSA group (P < 0.05). Levels of IL-4 in normal pregnancy group and URSA groups increased compared with the healthy control group, and the IL-4 levels in normal pregnancy group were significantly higher than those in URSA group (P < 0.05). Th1/Th2 imbalance, sTim-3 and Galectin-9 expression increase are found in the patients with URSA, ant this might be involved in the regulation of immunity in pregnancy.
Collapse
Affiliation(s)
- Mengru Wu
- Reproductive Medicine Center, First Affiliated Hospital of Xinjiang Medical UniversityUrumqi 830011, Xinjiang, P. R. China
| | - Yuejie Zhu
- Reproductive Medicine Center, First Affiliated Hospital of Xinjiang Medical UniversityUrumqi 830011, Xinjiang, P. R. China
| | - Jing Zhao
- Reproductive Medicine Center, First Affiliated Hospital of Xinjiang Medical UniversityUrumqi 830011, Xinjiang, P. R. China
| | - Haiquan Ai
- Reproductive Medicine Center, First Affiliated Hospital of Xinjiang Medical UniversityUrumqi 830011, Xinjiang, P. R. China
| | - Qiaoqiao Gong
- Reproductive Medicine Center, First Affiliated Hospital of Xinjiang Medical UniversityUrumqi 830011, Xinjiang, P. R. China
| | - Jia Zhang
- Reproductive Medicine Center, First Affiliated Hospital of Xinjiang Medical UniversityUrumqi 830011, Xinjiang, P. R. China
| | - Junda Zhao
- Reproductive Medicine Center, First Affiliated Hospital of Xinjiang Medical UniversityUrumqi 830011, Xinjiang, P. R. China
| | - Qingli Wang
- Reproductive Medicine Center, First Affiliated Hospital of Xinjiang Medical UniversityUrumqi 830011, Xinjiang, P. R. China
| | - Xiaoling La
- Reproductive Medicine Center, First Affiliated Hospital of Xinjiang Medical UniversityUrumqi 830011, Xinjiang, P. R. China
| | - Jianbing Ding
- Reproductive Medicine Center, First Affiliated Hospital of Xinjiang Medical UniversityUrumqi 830011, Xinjiang, P. R. China
- Department of Immunology, School of Preclinical Medicine of Xinjiang Medical UniversityUrumqi 830011, Xinjiang, P. R. China
| |
Collapse
|
39
|
Soyka MB, van de Veen W, Holzmann D, Akdis M, Akdis CA. Scientific foundations of allergen-specific immunotherapy for allergic disease. Chest 2015; 146:1347-1357. [PMID: 25367471 DOI: 10.1378/chest.14-0049] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Allergen-specific immunotherapy (AIT) was described as a therapeutic option for the treatment of allergies > 100 years ago. It is based on administration of allergen extracts and leads to the development of clinical allergen tolerance in selected patients. According to current knowledge, AIT results in the restoration of immune tolerance toward the allergen of interest. It is mainly accompanied by the induction of regulatory and suppressive subsets of T and B cells, the production of IgG4 isotype allergen-specific blocking antibodies, and decreased inflammatory responses to allergens by effector cells in inflamed tissues. Currently, AIT is mainly applied subcutaneously or sublingually and is suitable for both children and adults for pollen, pet dander, house dust mite, and venom allergies. It not only affects rhinoconjunctival symptoms but also has documented short- and long-term benefits in asthma treatment. Clinically, a fast onset of tolerance is achieved during desensitization, with a tolerable amount of side effects. The disease modification effect leads to decreased disease severity, less drug usage, prevention of future allergen sensitizations, and a long-term curative effect. Increasing safety while maintaining or even augmenting efficiency is the main goal of research for novel vaccine development and improvement of treatment schemes in AIT. This article reviews the principles of allergen-specific immune tolerance development and the effects of AIT in the clinical context.
Collapse
Affiliation(s)
- Michael B Soyka
- From the The Swiss Institute of Allergy and Asthma Research Davos, University of Zurich, Davos; Department of Otorhinolaryngology Head and Neck Surgery, University Hospital Zurich, Zurich
| | - Willem van de Veen
- From the The Swiss Institute of Allergy and Asthma Research Davos, University of Zurich, Davos
| | - David Holzmann
- Department of Otorhinolaryngology Head and Neck Surgery, University Hospital Zurich, Zurich
| | - Mübeccel Akdis
- From the The Swiss Institute of Allergy and Asthma Research Davos, University of Zurich, Davos
| | - Cezmi A Akdis
- From the The Swiss Institute of Allergy and Asthma Research Davos, University of Zurich, Davos; Department of Otorhinolaryngology Head and Neck Surgery, University Hospital Zurich, Zurich; Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland.
| |
Collapse
|
40
|
Fang XY, Xu WD, Pan HF, Leng RX, Ye DQ. Novel insights into Tim-4 function in autoimmune diseases. Autoimmunity 2014; 48:189-95. [DOI: 10.3109/08916934.2014.983266] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
41
|
Sinha S, Singh J, Jindal SK. Protective association of TIM1-1454G>a polymorphism with asthma in a North Indian population. Lung 2014; 193:31-8. [PMID: 25331534 DOI: 10.1007/s00408-014-9653-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 10/07/2014] [Indexed: 12/21/2022]
Abstract
PURPOSE TIM1 is a key regulator of Th2-dominated immune responses, including allergy, asthma, autoimmunity, and response to the pathogens. They are mainly expressed by hepatocytes and lymphoid cells. Analysis of the sequence of TIM1 was found to have range of SNPs which increases the transcriptional activity of the TIM1 gene. METHODS A case-control study was conducted with a total of 964 subjects, including 483 healthy controls and 481 asthma patients in the present study. DNA samples were extracted from blood, and genotyping was done using polymerase chain reaction-restriction fragment length polymorphism method. RESULTS Statistical analysis revealed that both heterozygous (GA) as well as the mutant (AA) genotype of -1454G>A (rs41297579) polymorphism shows resistance toward asthma with OR = 0.74, 95 % CI (0.55-0.98), p = 0.029 and OR = 0.43, 95 % CI (0.28-0.65), p = 0.000, respectively. The mutant (A) allele was also found to be highly protective toward asthma with OR = 0.68, 95 % CI (0.56-0.82) p = 0.000. However, no statistical difference was found between the TIM1-416G>C (rs9313422) polymorphism and asthma patients (p > 0.05). CONCLUSIONS This is the first study conducted in India conferring -1454G>A polymorphism provides resistance toward asthma while lack of association was found between -416G>C polymorphism and asthma in the studied North Indian population.
Collapse
Affiliation(s)
- Shweta Sinha
- Department of Biotechnology, Panjab University, Chandigarh, India
| | | | | |
Collapse
|
42
|
Cai XZ, Huang WY, Qiao Y, Chen Y, Du SY, Chen D, Yu S, Liu N, Dou LY, Jiang Y. Downregulation of TIM-3 mRNA expression in peripheral blood mononuclear cells from patients with systemic lupus erythematosus. ACTA ACUST UNITED AC 2014; 48:77-82. [PMID: 25493386 PMCID: PMC4288496 DOI: 10.1590/1414-431x20143701] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Accepted: 08/28/2014] [Indexed: 01/03/2023]
Abstract
The T-cell immunoglobulin and mucin domain (TIM) family is associated with autoimmune
diseases, but its expression level in the immune cells of systemic lupus
erythematosus (SLE) patients is not known. The aim of this study was to investigate
whether the expression of TIM-3 mRNA is associated with pathogenesis of SLE.
Quantitative real-time reverse transcription-polymerase chain reaction analysis
(qRT-PCR) was used to determine TIM-1, TIM-3, and TIM-4 mRNA expression in peripheral
blood mononuclear cells (PBMCs) from 132 patients with SLE and 62 healthy controls.
The PBMC surface protein expression of TIMs in PBMCs from 20 SLE patients and 15
healthy controls was assayed by flow cytometry. Only TIM-3 mRNA expression decreased
significantly in SLE patients compared with healthy controls (P<0.001). No
significant differences in TIM family protein expression were observed in leukocytes
from SLE patients and healthy controls (P>0.05). SLE patients with lupus nephritis
(LN) had a significantly lower expression of TIM-3 mRNA than those without LN
(P=0.001). There was no significant difference in the expression of TIM-3 mRNA within
different classes of LN (P>0.05). Correlation of TIM-3 mRNA expression with serum
IgA was highly significant (r=0.425, P=0.004), but was weakly correlated with total
serum protein (rs=0.283, P=0.049) and serum albumin (rs=0.297,
P=0.047). TIM-3 mRNA expression was weakly correlated with the Systemic Lupus
Erythematosus Disease Activity Index (SLEDAI; rs=-0.272, P=0.032). Our
results suggest that below-normal expression of TIM-3 mRNA in PBMC may be involved in
the pathogenesis of SLE.
Collapse
Affiliation(s)
- X Z Cai
- Central Laboratory, First Affiliated Hospital, China Medical University, Shenyang, China
| | - W Y Huang
- Central Laboratory, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Y Qiao
- Central Laboratory, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Y Chen
- Central Laboratory, First Affiliated Hospital, China Medical University, Shenyang, China
| | - S Y Du
- Central Laboratory, First Affiliated Hospital, China Medical University, Shenyang, China
| | - D Chen
- Central Laboratory, First Affiliated Hospital, China Medical University, Shenyang, China
| | - S Yu
- Central Laboratory, First Affiliated Hospital, China Medical University, Shenyang, China
| | - N Liu
- Department of Nephrology, First Affiliated Hospital, China Medical University, Shenyang, China
| | - L Y Dou
- Central Laboratory, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Y Jiang
- Central Laboratory, First Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
43
|
Elevated TIM3+ hematopoietic stem cells in untreated myelodysplastic syndrome displayed aberrant differentiation, overproliferation and decreased apoptosis. Leuk Res 2014; 38:714-21. [DOI: 10.1016/j.leukres.2014.03.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 03/02/2014] [Accepted: 03/22/2014] [Indexed: 11/24/2022]
|
44
|
Abstract
UNLABELLED T-cell immunoglobulin and mucin domain 1 (TIM-1) and other TIM family members were recently identified as phosphatidylserine (PtdSer)-mediated virus entry-enhancing receptors (PVEERs). These proteins enhance entry of Ebola virus (EBOV) and other viruses by binding PtdSer on the viral envelope, concentrating virus on the cell surface, and promoting subsequent internalization. The PtdSer-binding activity of the immunoglobulin-like variable (IgV) domain is essential for both virus binding and internalization by TIM-1. However, TIM-3, whose IgV domain also binds PtdSer, does not effectively enhance virus entry, indicating that other domains of TIM proteins are functionally important. Here, we investigate the domains supporting enhancement of enveloped virus entry, thereby defining the features necessary for a functional PVEER. Using a variety of chimeras and deletion mutants, we found that in addition to a functional PtdSer-binding domain PVEERs require a stalk domain of sufficient length, containing sequences that promote an extended structure. Neither the cytoplasmic nor the transmembrane domain of TIM-1 is essential for enhancing virus entry, provided the protein is still plasma membrane bound. Based on these defined characteristics, we generated a mimic lacking TIM sequences and composed of annexin V, the mucin-like domain of α-dystroglycan, and a glycophosphatidylinositol anchor that functioned as a PVEER to enhance transduction of virions displaying Ebola, Chikungunya, Ross River, or Sindbis virus glycoproteins. This identification of the key features necessary for PtdSer-mediated enhancement of virus entry provides a basis for more effective recognition of unknown PVEERs. IMPORTANCE T-cell immunoglobulin and mucin domain 1 (TIM-1) and other TIM family members are recently identified phosphatidylserine (PtdSer)-mediated virus entry-enhancing receptors (PVEERs). These proteins enhance virus entry by binding the phospholipid, PtdSer, present on the viral membrane. While it is known that the PtdSer binding is essential for the PVEER function of TIM-1, TIM-3 shares this binding activity but does not enhance virus entry. No comprehensive studies have been done to characterize the other domains of TIM-1. In this study, using a variety of chimeric proteins and deletion mutants, we define the features necessary for a functional PVEER. With these features in mind, we generated a TIM-1 mimic using functionally similar domains from other proteins. This mimic, like TIM-1, effectively enhanced transduction. These studies provide insight into the key features necessary for PVEERs and will allow for more effective identification of unknown PVEERs.
Collapse
|
45
|
Roth CG, Garner K, Eyck ST, Boyiadzis M, Kane LP, Craig FE. TIM3 expression by leukemic and non-leukemic myeloblasts. CYTOMETRY PART B-CLINICAL CYTOMETRY 2013; 84:167-72. [PMID: 23554257 DOI: 10.1002/cyto.b.21080] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 01/11/2013] [Accepted: 01/18/2013] [Indexed: 01/03/2023]
Abstract
BACKGROUND T-cell immunoglobulin mucin-3 (TIM3) has recently been described as an acute myeloid leukemia (AML) stem cell antigen expressed on leukemic myeloblasts, but not on normal hematopoietic stem cells. TIM3 is also expressed by monocytes, natural killer cells, and several T cell subsets; however, normal myeloblasts have not been well-characterized or compared to AML. A specific flow cytometric marker capable of separating leukemic myeloblasts from non-neoplastic myeloblasts would be diagnostically useful, especially in the post-chemotherapy setting. METHODS TIM3 myeloblast expression was assessed in 69 bone marrow and/or peripheral blood specimens, including 27 AML and 42 non-neoplastic cases (20 with a recent history of chemotherapy). TIM3 median fluorescence intensity (MFI) was evaluated within myeloblast, monocyte, T cell, and natural killer cell populations. RESULTS The median percentage of myeloblasts positive for TIM3 was lower in non-neoplastic specimens without a history of recent chemotherapy (50.3%) as compared to AML (71.4%), but not significantly different as compared to non-leukemic myeloblasts in the post-chemotherapy setting (72.4%). Mean myeloblast TIM3 MFI was higher in AML myeloblasts and non-leukemic myeloblasts in the post-chemotherapy setting as compared to non-neoplastic myeloblasts in cases lacking a history of chemotherapy. Mean monocyte, natural killer cell, and T-cell TIM3 MFI remained relatively constant in varied clinical settings. CONCLUSIONS We confirm that leukemic myeloblasts overexpress TIM3 as compared to non-neoplastic controls; however, high levels of expression may also be seen among non-leukemic myeloblasts in the post-chemotherapy setting. This overlap limits the diagnostic utility of TIM3 as a specific marker of neoplasia.
Collapse
Affiliation(s)
- Christine G Roth
- Department of Pathology, Division of Hematopathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Status of TIM-1 exon 4 haplotypes and CD4+T cell counts in HIV-1 seroprevalent North Indians. Hum Immunol 2013; 74:163-5. [DOI: 10.1016/j.humimm.2012.11.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 11/27/2012] [Accepted: 11/27/2012] [Indexed: 11/23/2022]
|
47
|
Soyer OU, Akdis M, Ring J, Behrendt H, Crameri R, Lauener R, Akdis CA. Mechanisms of peripheral tolerance to allergens. Allergy 2013; 68:161-70. [PMID: 23253293 DOI: 10.1111/all.12085] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2012] [Indexed: 12/24/2022]
Abstract
The immune system is regulated to protect the host from exaggerated stimulatory signals establishing a state of tolerance in healthy individuals. The disequilibrium in immune regulatory vs effector mechanisms results in allergic or autoimmune disorders in genetically predisposed subjects under certain environmental conditions. As demonstrated in allergen-specific immunotherapy and in the healthy immune response to high-dose allergen exposure models in humans, T regulatory cells are essential in the suppression of Th2-mediated inflammation, maintenance of immune tolerance, induction of the two suppressive cytokines interleukin-10 and transforming growth factor-β, inhibition of allergen-specific IgE, and enhancement of IgG4 and IgA. Also, suppression of dendritic cells, mast cells, and eosinophils contributes to the construction of peripheral tolerance to allergens. This review focuses on mechanisms of peripheral tolerance to allergens with special emphasis on recent developments in the area of immune regulation.
Collapse
Affiliation(s)
| | | | | | | | - R. Crameri
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zurich; Davos; Switzerland
| | | | | |
Collapse
|
48
|
Fierro NA, Castro-Garcia FP, Panduro A. Rethinking cytokine function during hepatitis A and hepatitis C infections. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/abb.2013.47a1003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|