1
|
Abooali M, Schlichtner S, Lei X, Aliu N, Ruggiero S, Loges S, Ziegler M, Hertel F, Volckmar AL, Stenzinger A, Christopoulos P, Thomas M, Klenova E, Meyer NH, Boussios S, Heaton N, Zen Y, Zamalloa A, Chokshi S, Urbani L, Richard S, Kirubendran K, Hussain R, Siligardi G, Cholewa D, Berger SM, Yasinska IM, Fasler-Kan E, Sumbayev VV. Intracellular and extracellular activities of V-domain Ig-containing suppressor of T cell activation (VISTA) modulated by immunosuppressive factors of tumour microenvironment. Cancer Lett 2025; 616:217581. [PMID: 39983894 DOI: 10.1016/j.canlet.2025.217581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 02/23/2025]
Abstract
V-domain Ig-containing suppressor of T cell activation (VISTA) is a unique immune checkpoint protein, which was reported to display both receptor and ligand activities. However, the mechanisms of regulation of VISTA activity and functions by factors of tumour microenvironment (TME) remain unclear and understanding these processes is required in order to develop successful personalised cancer immunotherapeutic strategies and approaches. Here we report for the very first time that VISTA interacts with another immune checkpoint protein galectin-9 inside the cell most likely facilitating its interaction with TGF-β-activated kinase 1 (TAK1). This process is required for protection of lysosomes, which is crucial for many cell types and tissues. We found that VISTA expression can be differentially controlled by crucial factors present in TME, such as transforming growth factor beta type 1 (TGF-β) and hypoxia as well as other factors activating hypoxic signalling. We confirmed that involvement of these important pathways modulated by TME differentially influences VISTA expression in different cell types. These networks include: TGF-β-Smad3 pathway, TAK1 (TGF-β-activated kinase 1) or apoptosis signal-regulating kinase 1 (ASK1)-induced activation of activating transcription factor 2 (ATF-2) and hypoxic signalling pathway. Based on this work we determined the five critical functions of VISTA and the role of TME factors in controlling (modulating or downregulating) VISTA expression.
Collapse
Affiliation(s)
- Maryam Abooali
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, UK
| | - Stephanie Schlichtner
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Xi Lei
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, UK
| | - Nijas Aliu
- Department of Human Genetics, Children's Hospital, Inselspital, University of Bern, Bern, Switzerland
| | - Sabrina Ruggiero
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Sonia Loges
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Martin Ziegler
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Franziska Hertel
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Anna-Lena Volckmar
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Translational Lung Research Center (TLRC) Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik, Heidelberg University Hospital, Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Michael Thomas
- Department of Thoracic Oncology, Thoraxklinik, Heidelberg University Hospital, Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Elena Klenova
- School of Life Sciences, University of Essex, Colchester, UK
| | - N Helge Meyer
- Division of General and Visceral Surgery, Department of Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Stergios Boussios
- Faculty of Medicine, Health and Social Care, Canterbury Christ Church University, Canterbury, CT1 1QU, Kent, UK; Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King's College London, Strand, London, WC2R 2LS, UK; Kent Medway Medical School, University of Kent, Canterbury, CT2 7LX, Kent, UK; Department of Medical Oncology, Medway NHS Foundation Trust, Gillingham, ME7 5NY, Kent, UK; AELIA Organization, 9th Km Thessaloniki-Thermi, 57001, Thessaloniki, Greece
| | - Nigel Heaton
- Institute of Liver Studies, King's College Hospital, Denmark Hill, London, UK
| | - Yoh Zen
- Institute of Liver Studies, King's College Hospital, Denmark Hill, London, UK
| | - Ane Zamalloa
- Institute of Liver Studies, King's College Hospital, Denmark Hill, London, UK
| | - Shilpa Chokshi
- Peninsula Medical School, Faculty of Health, University of Plymouth, UK; Roger Williams Institute of Liver Studies, School of Immunology & Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, Foundation for Liver Research and King's College Hospital, London, UK
| | - Luca Urbani
- Roger Williams Institute of Liver Studies, School of Immunology & Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, Foundation for Liver Research and King's College Hospital, London, UK
| | - Sophie Richard
- Roger Williams Institute of Liver Studies, School of Immunology & Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, Foundation for Liver Research and King's College Hospital, London, UK
| | - Kavitha Kirubendran
- Roger Williams Institute of Liver Studies, School of Immunology & Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, Foundation for Liver Research and King's College Hospital, London, UK
| | | | | | - Dietmar Cholewa
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Steffen M Berger
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Inna M Yasinska
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, UK
| | - Elizaveta Fasler-Kan
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland.
| | - Vadim V Sumbayev
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, UK.
| |
Collapse
|
2
|
Behranvand N, Nasri F, Moghaddam MV, Gholami MD, Safari E, Esmaeil N, Falak R. Galectin-9 induces IL-1β production as a key inflammatory cytokine in the acute myeloid leukemia cell line (U937). Res Pharm Sci 2025; 20:304-315. [PMID: 40444162 PMCID: PMC12118778 DOI: 10.4103/rps.rps_234_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 06/02/2025] Open
Abstract
Background and purpose T-cell immunoglobulin and mucin-domain containing protein-3 (TIM-3)/ galectin-9 (Gal-9)/ autocrine loop in myeloid leukemia stem cells provokes inflammation through the NF-κB signaling pathway, which is influential in the expression of inflammatory factors. Interleukin 1β (IL-1β) is a vital inflammatory cytokine that plays an important role in the proliferation and therapy resistance of acute myeloid leukemia (AML) cells. This study aimed to assess the effect of Gal-9 on IL-1β in the human leukemic U937 cell line. Experimental approach The U937 cells were cultured in different concentrations of Gal-9. Cell counting kit-8 was used to assess the effect of Gal-9 on human leukemic U937 cell proliferation. Also, its impact on the expression of TIM-3, Gal-9, IL-1β, IL-1βR, IL-1βRAP, and NLRP3 genes and IL-1β protein was studied by RT-PCR and ELISA, respectively. Moreover, the effect of Gal-9 on the NF-κB signaling pathway was evaluated by western blotting. Findings/Results U937 cells were expanded in the presence of Gal-9 in a concentration-dependent manner. Following treatment of U937 cells with Gal-9, the gene expression of Gal-9, IL-1B, IL-1BR, and IL-1BRAP were significantly upregulated compared to the control group. The IL-1β concentration increased following Gal-9 treatment in a concentration-dependent manner, while following time-pass its level significantly decreased. Furthermore, Gal-9 slightly increased NF-κB phosphorylation. Conclusion and implications Gal-9 increased IL-1β level as a critical inflammatory cytokine in the proliferation and resistance of AML cells to therapy. According to this finding, targeting and blocking the TIM-3/Gal-9 autocrine loop can suppress IL-1β production and facilitate AML treatment.
Collapse
Affiliation(s)
- Nafiseh Behranvand
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farzad Nasri
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Davoodzadeh Gholami
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elahe Safari
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nafiseh Esmaeil
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Falak
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Heidari F, Kazemi-Sefat NA, Feizollahi P, Gerdabi S, Pourfathollah AA, Ebtekar M. Effect of FLT3 ligand on the gene expression of TIM-3, HIF1-α, and TNF-α in an acute myeloid leukemia cell line. Mol Biol Rep 2025; 52:313. [PMID: 40085277 DOI: 10.1007/s11033-025-10396-4] [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: 12/30/2024] [Accepted: 02/26/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Acute Myeloid Leukemia (AML) pathogenesis is driven by the dysregulation of various cell signaling pathways, including the FMS-Like Tyrosine Kinase 3 (FLT3) pathway and its ligand (FLT3L). These pathways play a critical role in promoting cell survival, proliferation, and resistance to apoptosis, contributing to leukemogenesis. In this study, we investigated the effects of FLT3L on the expression of key genes associated with immune regulation, hypoxia, and inflammation-TIM-3, HIF-1α, and TNF-α-in the THP-1 cell line, a well-established model for AML research. METHODS THP-1 cells were cultured under standard conditions and treated with varying concentrations of FLT3L, alongside PMA as a positive control. Quantitative RT-PCR was employed to measure the expression levels of TIM-3, HIF-1α, and TNF-α genes after 48 h of treatment. RESULTS Our findings demonstrated that specific concentrations of FLT3L significantly upregulated the expression of TIM-3, HIF-1α, and TNF-α in THP-1 cells. This suggests that FLT3L not only influences cell proliferation and survival but also modulates pathways related to immune evasion, hypoxia adaptation, and inflammatory responses, which are hallmarks of leukemia progression. CONCLUSION These results highlight the pivotal role of FLT3L in regulating the expression of genes associated with AML pathogenesis, particularly those involved in hypoxia (HIF-1α), immune checkpoint regulation (TIM-3), and inflammation (TNF-α). The findings underscore the potential of targeting the FLT3 pathway as a therapeutic strategy in AML. Further studies are warranted to elucidate the underlying molecular mechanisms and explore their clinical implications for improving patient outcomes.
Collapse
Affiliation(s)
- Fatemeh Heidari
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 14155-114, Iran
| | - Nazanin Atieh Kazemi-Sefat
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 14155-114, Iran
| | - Parisa Feizollahi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 14155-114, Iran
| | - Sajjad Gerdabi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 14155-114, Iran
| | - Ali Akbar Pourfathollah
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 14155-114, Iran
| | - Masoumeh Ebtekar
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 14155-114, Iran.
| |
Collapse
|
4
|
Luo J, Zhang C, Chen D, Chang T, Chen S, Lin Z, Yi C, Tang ZH. Tim-3 pathway dysregulation and targeting in sepsis-induced immunosuppression. Eur J Med Res 2024; 29:583. [PMID: 39696711 PMCID: PMC11656820 DOI: 10.1186/s40001-024-02203-w] [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: 07/18/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024] Open
Abstract
Sepsis is a major medical problem which causes millions of deaths worldwide every year. The host immune response in sepsis is characterized by acute inflammation and a simultaneous state of immunosuppression. In the later stage of sepsis, immunosuppression is a crucial factor that increases the susceptibility of septic patients to secondary infection and mortality. It is characterized by T cell exhaustion, excessive production of anti-inflammatory cytokines, hyperproliferation of immune suppressor cells and aberrant expression of immune checkpoint molecules. T cell immunoglobulin and mucin domain 3 (Tim-3), an immune checkpoint molecule, is found on the surface of various cells, including macrophages, NK cells, NKT cells, and T cells. There are four different ligands for Tim-3, and accumulating evidence indicates that Tim-3 and its ligands play a crucial role in regulating immune cell dysfunction during sepsis. Anti-Tim-3 antibodies have been applied in the field of cancer immunotherapy and have achieved positive therapeutic effects in some clinical trials. However, the therapeutic efficacy of Tim-3 blockade is still controversial in animal models of sepsis. These challenges highlight the need for a deeper understanding of Tim-3 signaling in sepsis. This review examines the comprehensive effect of Tim-3 signaling in the development of sepsis-induced immunosuppression and the therapeutic efficacy of Tim-3 blockade.
Collapse
Affiliation(s)
- Jialiu Luo
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong Zhang
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Deng Chen
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Teding Chang
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shunyao Chen
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Lin
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengla Yi
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Zhao-Hui Tang
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
5
|
Mendoza M, Ballesteros A, Rendon-Correa E, Tonk R, Warren J, Snow AL, Stowell SR, Blois SM, Dveksler G. Modulation of galectin-9 mediated responses in monocytes and T-cells by pregnancy-specific glycoprotein 1. J Biol Chem 2024; 300:107638. [PMID: 39121996 PMCID: PMC11403483 DOI: 10.1016/j.jbc.2024.107638] [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: 05/20/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
Successful pregnancy relies on a coordinated interplay between endocrine, immune, and metabolic processes to sustain fetal growth and development. The orchestration of these processes involves multiple signaling pathways driving cell proliferation, differentiation, angiogenesis, and immune regulation necessary for a healthy pregnancy. Among the molecules supporting placental development and maternal tolerance, the families of pregnancy-specific glycoproteins and galectins are of great interest in reproductive biology. We previously found that PSG1 can bind to galectin-1 (GAL-1). Herein, we characterized the interaction between PSG1 and other members of the galectin family expressed during pregnancy, including galectin-3, -7, -9, and -13 (GAL-3, GAL-7, GAL-9, and GAL-13). We observed that PSG1 binds to GAL-1, -3, and -9, with the highest apparent affinity seen for GAL-9, and that the interaction of PSG1 with GAL-9 is carbohydrate-dependent. We further investigated the ability of PSG1 to regulate GAL-9 responses in human monocytes, a murine macrophage cell line, and T-cells, and determined whether PSG1 binds to both carbohydrate recognition domains of GAL-9. Additionally, we compared the apparent affinity of GAL-9 binding to PSG1 with other known GAL-9 ligands in these cells, Tim-3 and CD44. Lastly, we explored functional conservation between murine and human PSGs by determining that Psg23, a highly expressed member of the murine Psg family, can bind some murine galectins despite differences in amino acid composition and domain structure.
Collapse
Affiliation(s)
- Mirian Mendoza
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Angela Ballesteros
- Section on Sensory Physiology and Biophysics, National Institute on Deafness and other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA
| | - Elizabeth Rendon-Correa
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Rohan Tonk
- Section on Sensory Physiology and Biophysics, National Institute on Deafness and other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA
| | - James Warren
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Andrew L Snow
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Sean R Stowell
- Department of Pathology, Brigham and Women's Hospital, Boston Massachusetts, USA
| | - Sandra M Blois
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Glyco-HAM, a cooperation of Universität Hamburg, Technology Platform Mass Spectrometry and University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gabriela Dveksler
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.
| |
Collapse
|
6
|
Yıldırım C. Galectin-9, a pro-survival factor inducing immunosuppression, leukemic cell transformation and expansion. Mol Biol Rep 2024; 51:571. [PMID: 38662155 DOI: 10.1007/s11033-024-09563-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 04/17/2024] [Indexed: 04/26/2024]
Abstract
Leukemia is a malignancy of the bone marrow and blood originating from self-renewing cancerous immature blast cells or transformed leukocytes. Despite improvements in treatments, leukemia remains still a serious disease with poor prognosis because of disease heterogeneity, drug resistance and relapse. There is emerging evidence that differentially expression of co-signaling molecules play a critical role in tumor immune evasion. Galectin-9 (Gal-9) is one of the key proteins that leukemic cells express, secrete, and use to proliferate, self-renew, and survive. It also suppresses host immune responses controlled by T and NK cells, enabling leukemic cells to evade immune surveillance. The present review provides the molecular mechanisms of Gal-9-induced immune evasion in leukemia. Understanding the complex immune evasion machinery driven by Gal-9 expressing leukemic cells will enable the identification of novel therapeutic strategies for efficient immunotherapy in leukemic patients. Combined treatment approaches targeting T-cell immunoglobulin and mucin domain-3 (Tim-3)/Gal-9 and other immune checkpoint pathways can be considered, which may enhance the efficacy of host effector cells to attack leukemic cells.
Collapse
Affiliation(s)
- Cansu Yıldırım
- Atatürk Vocational School of Health Services, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey.
| |
Collapse
|
7
|
Shapourian H, Ghanadian M, Eskandari N, Shokouhi A, Demirel GY, Bazhin AV, Ganjalikhani-Hakemi M. TIM-3/Galectin-9 interaction and glutamine metabolism in AML cell lines, HL-60 and THP-1. BMC Cancer 2024; 24:125. [PMID: 38267906 PMCID: PMC10809689 DOI: 10.1186/s12885-024-11898-3] [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: 10/22/2023] [Accepted: 01/19/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND T cell immunoglobulin and mucin-domain containing-3 (TIM-3) is a cell surface molecule that was first discovered on T cells. However, recent studies revealed that it is also highly expressed in acute myeloid leukemia (AML) cells and it is related to AML progression. As, Glutamine appears to play a prominent role in malignant tumor progression, especially in their myeloid group, therefore, in this study we aimed to evaluate the relation between TIM-3/Galectin-9 axis and glutamine metabolism in two types of AML cell lines, HL-60 and THP-1. METHODS Cell lines were cultured in RPMI 1640 which supplemented with 10% FBS and 1% antibiotics. 24, 48, and 72 h after addition of recombinant Galectin-9 (Gal-9), RT-qPCR analysis, RP-HPLC and gas chromatography techniques were performed to evaluate the expression of glutaminase (GLS), glutamate dehydrogenase (GDH) enzymes, concentration of metabolites; Glutamate (Glu) and alpha-ketoglutarate (α-KG) in glutaminolysis pathway, respectively. Western blotting and MTT assay were used to detect expression of mammalian target of rapamycin complex (mTORC) as signaling factor, GLS protein and cell proliferation rate, respectively. RESULTS The most mRNA expression of GLS and GDH in HL-60 cells was seen at 72 h after Gal-9 treatment (p = 0.001, p = 0.0001) and in THP-1 cell line was observed at 24 h after Gal-9 addition (p = 0.001, p = 0.0001). The most mTORC and GLS protein expression in HL-60 and THP-1 cells was observed at 72 and 24 h after Gal-9 treatment (p = 0.0001), respectively. MTT assay revealed that Gal-9 could promote cell proliferation rate in both cell lines (p = 0.001). Glu concentration in HL-60 and α-KG concentration in both HL-60 (p = 0.03) and THP-1 (p = 0.0001) cell lines had a decreasing trend. But, Glu concentration had an increasing trend in THP-1 cell line (p = 0.0001). CONCLUSION Taken together, this study suggests TIM-3/Gal-9 interaction could promote glutamine metabolism in HL-60 and THP-1 cells and resulting in AML development.
Collapse
Affiliation(s)
- Hooriyeh Shapourian
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mustafa Ghanadian
- Department of Pharmacognosy, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nahid Eskandari
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abolfazl Shokouhi
- Department of Endocrine and metabolism research center, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Alexandr V Bazhin
- Department of General, Visceral and Transplant Surgery, Ludwig Maximilians University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Mazdak Ganjalikhani-Hakemi
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey.
| |
Collapse
|
8
|
Abooali M, Yasinska IM, Schlichtner S, Ruggiero S, Berger SM, Cholewa D, Milošević M, Bartenstein A, Fasler-Kan E, Sumbayev VV. Activation of immune evasion machinery is a part of the process of malignant transformation of human cells. Transl Oncol 2024; 39:101805. [PMID: 37844478 PMCID: PMC10587773 DOI: 10.1016/j.tranon.2023.101805] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/02/2023] [Accepted: 10/09/2023] [Indexed: 10/18/2023] Open
Abstract
Malignant transformation of human cells is associated with their re-programming which results in uncontrolled proliferation and in the same time biochemical activation of immunosuppressive pathways which form cancer immune evasion machinery. However, there is no conceptual understanding of whether immune evasion machinery pathways and expression of immune checkpoint proteins form a part of the process of malignant transformation or if they are triggered by T lymphocytes and natural killers (NK) attempting to attack cells which are undergoing or already underwent malignant transformation. To address this fundamental question, we performed experimental malignant transformation of BEAS-2B human bronchial epithelium cells and RC-124 non-malignant human kidney epithelial cells using bracken extracts containing carcinogenic alkaloid called ptaquiloside. This transformation led to a significant upregulation of cell proliferation velocity and in the same time led to a significant upregulation in expression of key immune checkpoint proteins - galectin-9, programmed death ligand 1 (PD-L1), indoleamine 2,3-dioxygenase (IDO1). Their increased expression levels were in line with upregulation of the levels and activities of HIF-1 transcription complex and transforming growth factor beta type 1 (TGF-β)-Smad3 signalling pathway. When co-cultured with T cells, transformed epithelial cells displayed much higher and more efficient immune evasion activity compared to original non-transformed cells. Therefore, this work resolved a very important scientific and clinical question and suggested that cancer immune evasion machinery is activated during malignant transformation of human cells regardless the presence of immune cells in microenvironment.
Collapse
Affiliation(s)
- Maryam Abooali
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - Inna M Yasinska
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - Stephanie Schlichtner
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom; DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ); German Center for Lung Research (DZL), Heidelberg, Germany; Department of Personalized Oncology, Medical Faculty Mannheim, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| | - Sabrina Ruggiero
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Steffen M Berger
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Dietmar Cholewa
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Milan Milošević
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Andreas Bartenstein
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Elizaveta Fasler-Kan
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland.
| | - Vadim V Sumbayev
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom.
| |
Collapse
|
9
|
Rezaei M, Ghanadian M, Ghezelbash B, Shokouhi A, Bazhin AV, Zamyatnin AA, Ganjalikhani-Hakemi M. TIM-3/Gal-9 interaction affects glucose and lipid metabolism in acute myeloid leukemia cell lines. Front Immunol 2023; 14:1267578. [PMID: 38022614 PMCID: PMC10667689 DOI: 10.3389/fimmu.2023.1267578] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction T-cell immunoglobulin and mucin domain-3 (TIM-3) is a transmembrane molecule first identified as an immunoregulator. This molecule is also expressed on leukemic cells in acute myeloid leukemia and master cell survival and proliferation. In this study, we aimed to explore the effect of TIM-3 interaction with its ligand galectin-9 (Gal-9) on glucose and lipid metabolism in AML cell lines. Methods HL-60 and THP-1 cell lines, representing M3 and M5 AML subtypes, respectively, were cultured under appropriate conditions. The expression of TIM-3 on the cell surface was ascertained by flow cytometric assay. We used real-time PCR to examine the mRNA expression of GLUT-1, HK-2, PFKFB-3, G6PD, ACC-1, ATGL, and CPT-1A; colorimetric assays to measure the concentration of glucose, lactate, GSH, and the enzymatic activity of G6PD; MTT assay to determine cellular proliferation; and gas chromatography-mass spectrometry (GC-MS) to designate FFAs. Results We observed the significant upregulated expression of GLUT-1, HK-2, PFKFB-3, ACC-1, CPT-1A, and G6PD and the enzymatic activity of G6PD in a time-dependent manner in the presence of Gal-9 compared to the PMA and control groups in both HL-60 and THP-1 cell lines (p > 0.05). Moreover, the elevation of extracellular free fatty acids, glucose consumption, lactate release, the concentration of cellular glutathione (GSH) and cell proliferation were significantly higher in the presence of Gal-9 compared to the PMA and control groups in both cell lines (p < 0.05). Conclusion TIM-3/Gal-9 ligation on AML cell lines results in aerobic glycolysis and altered lipid metabolism and also protects cells from oxidative stress, all in favor of leukemic cell survival and proliferation.
Collapse
Affiliation(s)
- Mahnaz Rezaei
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mustafa Ghanadian
- Department of Pharmacognosy, School of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Behrooz Ghezelbash
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abolfazl Shokouhi
- Endocrine and Metabolism Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alexandr V. Bazhin
- Department of General, Visceral and Transplant Surgery, Ludwig Maximilians University of Munich, Munich, Germany
| | - Andrey A. Zamyatnin
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, Sochi, Russia
- Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Mazdak Ganjalikhani-Hakemi
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Türkiye
| |
Collapse
|
10
|
Ning Q, Jian T, Cui S, Shi L, Jian X, He X, Zhang X, Li X. Tim-3 facilitates immune escape in benzene-induced acute myeloid leukemia mouse model by promoting macrophage M2 polarization. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 266:115532. [PMID: 37806131 DOI: 10.1016/j.ecoenv.2023.115532] [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: 06/19/2023] [Revised: 09/11/2023] [Accepted: 09/26/2023] [Indexed: 10/10/2023]
Abstract
Benzene poisoning can cause acute myeloid leukemia (AML) through a variety of passways. Tim-3 has gained prominence as a potential candidate in mediating immunosuppression in tumor microenvironments. The macrophage polarization is also related to immune escape. Herein, we reported that Tim-3 and macrophage M2 polarization play a vital role in benzene-induced AML. First, the benzene-induced AML C3H/He mouse model was constructed by subcutaneously injecting 250 mg/kg of benzene. After six months, macrophage phenotype, cytokines, and Tim-3 expression levels were investigated. Flow cytometry assay revealed that the T-cell inhibitory receptor Tim-3 was significantly upregulated in both bone marrow and spleen of the benzene-induced AML mouse model. Elisa's results displayed a decreased serum level of IL-12 while increased TGF-β1. Mechanistically, changes in cytokine secretion promote the growth of M2-type macrophages in the bone marrow and spleen, as determined by immunofluorescence assay. The increased levels of PI3K, AKT, and mTOR in the benzene-exposure group further proved the crucial role of Tim-3 in regulating the functional status of macrophages in the AML microenvironment. These results demonstrate that Tim-3 and macrophage polarization may play a vital role during the immune escape of the benzene-induced AML. This study provides a new potential intervention site for immune checkpoint-based AML therapeutic strategy.
Collapse
Affiliation(s)
- Qiong Ning
- Department of Occupational Diseases, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250002, China
| | - Tianzi Jian
- Department of Poisoning and Occupational Diseases, Emergency Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Siqi Cui
- Department of Poisoning and Occupational Diseases, Emergency Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Longke Shi
- Department of Poisoning and Occupational Diseases, Emergency Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiangdong Jian
- Department of Poisoning and Occupational Diseases, Emergency Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiaopeng He
- Department of Thoracic surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Xiangxing Zhang
- Department of Occupational and Environmental Health, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiangxin Li
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
11
|
Bakhtiyari M, Liaghat M, Aziziyan F, Shapourian H, Yahyazadeh S, Alipour M, Shahveh S, Maleki-Sheikhabadi F, Halimi H, Forghaniesfidvajani R, Zalpoor H, Nabi-Afjadi M, Pornour M. The role of bone marrow microenvironment (BMM) cells in acute myeloid leukemia (AML) progression: immune checkpoints, metabolic checkpoints, and signaling pathways. Cell Commun Signal 2023; 21:252. [PMID: 37735675 PMCID: PMC10512514 DOI: 10.1186/s12964-023-01282-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/17/2023] [Indexed: 09/23/2023] Open
Abstract
Acute myeloid leukemia (AML) comprises a multifarious and heterogeneous array of illnesses characterized by the anomalous proliferation of myeloid cells in the bone marrow microenvironment (BMM). The BMM plays a pivotal role in promoting AML progression, angiogenesis, and metastasis. The immune checkpoints (ICs) and metabolic processes are the key players in this process. In this review, we delineate the metabolic and immune checkpoint characteristics of the AML BMM, with a focus on the roles of BMM cells e.g. tumor-associated macrophages, natural killer cells, dendritic cells, metabolic profiles and related signaling pathways. We also discuss the signaling pathways stimulated in AML cells by BMM factors that lead to AML progression. We then delve into the roles of immune checkpoints in AML angiogenesis, metastasis, and cell proliferation, including co-stimulatory and inhibitory ICs. Lastly, we discuss the potential therapeutic approaches and future directions for AML treatment, emphasizing the potential of targeting metabolic and immune checkpoints in AML BMM as prognostic and therapeutic targets. In conclusion, the modulation of these processes through the use of directed drugs opens up new promising avenues in combating AML. Thereby, a comprehensive elucidation of the significance of these AML BMM cells' metabolic and immune checkpoints and signaling pathways on leukemic cells can be undertaken in the future investigations. Additionally, these checkpoints and cells should be considered plausible multi-targeted therapies for AML in combination with other conventional treatments in AML. Video Abstract.
Collapse
Affiliation(s)
- Maryam Bakhtiyari
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Mahsa Liaghat
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Medical Laboratory Sciences, Faculty of Medical Sciences, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Fatemeh Aziziyan
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hooriyeh Shapourian
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sheida Yahyazadeh
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maedeh Alipour
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Shaghayegh Shahveh
- American Association of Naturopath Physician (AANP), Washington, DC, USA
| | - Fahimeh Maleki-Sheikhabadi
- Department of Hematology and Blood Banking, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Halimi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Razieh Forghaniesfidvajani
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Hamidreza Zalpoor
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Majid Pornour
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA.
| |
Collapse
|
12
|
Schlichtner S, Yasinska IM, Klenova E, Abooali M, Lall GS, Berger SM, Ruggiero S, Cholewa D, Milošević M, Gibbs BF, Fasler-Kan E, Sumbayev VV. L-Kynurenine participates in cancer immune evasion by downregulating hypoxic signaling in T lymphocytes. Oncoimmunology 2023; 12:2244330. [PMID: 37577144 PMCID: PMC10416736 DOI: 10.1080/2162402x.2023.2244330] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/23/2023] [Accepted: 07/31/2023] [Indexed: 08/15/2023] Open
Abstract
Malignant tumors often escape anticancer immune surveillance by suppressing the cytotoxic functions of T lymphocytes. While many of these immune evasion networks include checkpoint proteins, small molecular weight compounds, such as the amino acid L-kynurenine (LKU), could also substantially contribute to the suppression of anti-cancer immunity. However, the biochemical mechanisms underlying the suppressive effects of LKU on T-cells remain unclear. Here, we report for the first time that LKU suppresses T cell function as an aryl hydrocarbon receptor (AhR) ligand. The presence of LKU in T cells is associated with AhR activation, which results in competition between AhR and hypoxia-inducible factor 1 alpha (HIF-1α) for the AhR nuclear translocator, ARNT, leading to T cell exhaustion. The expression of indoleamine 2,3-dioxygenase 1 (IDO1, the enzyme that leads to LKU generation) is induced by the TGF-β-Smad-3 pathway. We also show that IDO-negative cancers utilize an alternative route for LKU production via the endogenous inflammatory mediator, the high mobility group box 1 (HMGB-1)-interferon-gamma (IFN-γ) axis. In addition, other IDO-negative tumors (like T-cell lymphomas) trigger IDO1 activation in eosinophils present in the tumor microenvironment (TME). These mechanisms suppress cytotoxic T cell function, and thus support the tumor immune evasion machinery.
Collapse
Affiliation(s)
- Stephanie Schlichtner
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, UK
- Department of Personalized Medical Oncology, DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ); German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Personalized Oncology, University Hospital Mannheim, Mannheim, Germany
| | - Inna M. Yasinska
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, UK
| | - Elena Klenova
- School of Biological Sciences, University of Essex, Colchester, UK
| | - Maryam Abooali
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, UK
| | - Gurprit S. Lall
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, UK
| | - Steffen M. Berger
- Department of Pediatric Surgery, Children’s Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Sabrina Ruggiero
- Department of Pediatric Surgery, Children’s Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland
- Department of Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Dietmar Cholewa
- Department of Pediatric Surgery, Children’s Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Milan Milošević
- Department of Pediatric Surgery, Children’s Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Bernhard F. Gibbs
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, UK
- Department of Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Elizaveta Fasler-Kan
- Department of Pediatric Surgery, Children’s Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Vadim V. Sumbayev
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, UK
| |
Collapse
|
13
|
Astore S, Baciarello G, Cerbone L, Calabrò F. Primary and acquired resistance to first-line therapy for clear cell renal cell carcinoma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:517-546. [PMID: 37842234 PMCID: PMC10571064 DOI: 10.20517/cdr.2023.33] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/26/2023] [Accepted: 07/11/2023] [Indexed: 10/17/2023]
Abstract
The introduction of first-line combinations had improved the outcomes for metastatic renal cell carcinoma (mRCC) compared to sunitinib. However, some patients either have inherent resistance or develop resistance as a result of the treatment. Depending on the kind of therapy employed, many factors underlie resistance to systemic therapy. Angiogenesis and the tumor immune microenvironment (TIME), nevertheless, are inextricably linked. Although angiogenesis and the manipulation of the tumor microenvironment are linked to hypoxia, which emerges as a hallmark of renal cell carcinoma (RCC) pathogenesis, it is only one of the potential elements involved in the distinctive intra- and inter-tumor heterogeneity of RCC that is still dynamic. We may be able to more correctly predict therapy response and comprehend the mechanisms underlying primary or acquired resistance by integrating tumor genetic and immunological markers. In order to provide tools for patient selection and to generate hypotheses for the development of new strategies to overcome resistance, we reviewed the most recent research on the mechanisms of primary and acquired resistance to immune checkpoint inhibitors (ICIs) and tyrosine kinase inhibitors (TKIs) that target the vascular endothelial growth factor receptor (VEGFR).We can choose patients' treatments and cancer preventive strategies using an evolutionary approach thanks to the few evolutionary trajectories that characterize ccRCC.
Collapse
Affiliation(s)
- Serena Astore
- Medical Oncology, San Camillo Forlanini Hospital, Rome 00152, Italy
| | | | - Linda Cerbone
- Medical Oncology, San Camillo Forlanini Hospital, Rome 00152, Italy
| | - Fabio Calabrò
- Medical Oncology, San Camillo Forlanini Hospital, Rome 00152, Italy
- Medical Oncology, IRCSS, National Cancer Institute Regina Elena, Rome 00128, Italy
| |
Collapse
|
14
|
Benito-Lopez JJ, Marroquin-Muciño M, Perez-Medina M, Chavez-Dominguez R, Aguilar-Cazares D, Galicia-Velasco M, Lopez-Gonzalez JS. Partners in crime: The feedback loop between metabolic reprogramming and immune checkpoints in the tumor microenvironment. Front Oncol 2023; 12:1101503. [PMID: 36713558 PMCID: PMC9879362 DOI: 10.3389/fonc.2022.1101503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/22/2022] [Indexed: 01/15/2023] Open
Abstract
The tumor microenvironment (TME) is a complex and constantly changing cellular system composed of heterogeneous populations of tumor cells and non-transformed stromal cells, such as stem cells, fibroblasts, endothelial cells, pericytes, adipocytes, and innate and adaptive immune cells. Tumor, stromal, and immune cells consume available nutrients to sustain their proliferation and effector functions and, as a result of their metabolism, produce a wide array of by-products that gradually alter the composition of the milieu. The resulting depletion of essential nutrients and enrichment of by-products work together with other features of the hostile TME to inhibit the antitumor functions of immune cells and skew their phenotype to promote tumor progression. This review briefly describes the participation of the innate and adaptive immune cells in recognizing and eliminating tumor cells and how the gradual metabolic changes in the TME alter their antitumor functions. In addition, we discuss the overexpression of the immune checkpoints and their ligands as a result of nutrient deprivation and by-products accumulation, as well as the amplification of the metabolic alterations induced by the immune checkpoints, which creates an immunosuppressive feedback loop in the TME. Finally, the combination of metabolic and immune checkpoint inhibitors as a potential strategy to treat cancer and enhance the outcome of patients is highlighted.
Collapse
Affiliation(s)
- Jesus J Benito-Lopez
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosio Villegas", Mexico City, Mexico
- Posgrado en Ciencias Biologicas, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Mario Marroquin-Muciño
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosio Villegas", Mexico City, Mexico
- Laboratorio de Quimioterapia Experimental, Departamento de Bioquimica, Escuela Nacional de Ciencias Biologicas, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Mario Perez-Medina
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosio Villegas", Mexico City, Mexico
- Laboratorio de Quimioterapia Experimental, Departamento de Bioquimica, Escuela Nacional de Ciencias Biologicas, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Rodolfo Chavez-Dominguez
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosio Villegas", Mexico City, Mexico
- Posgrado en Ciencias Biologicas, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Dolores Aguilar-Cazares
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosio Villegas", Mexico City, Mexico
| | - Miriam Galicia-Velasco
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosio Villegas", Mexico City, Mexico
| | - Jose S Lopez-Gonzalez
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosio Villegas", Mexico City, Mexico
| |
Collapse
|
15
|
Schlichtner S, Yasinska IM, Lall GS, Berger SM, Ruggiero S, Cholewa D, Aliu N, Gibbs BF, Fasler-Kan E, Sumbayev VV. T lymphocytes induce human cancer cells derived from solid malignant tumors to secrete galectin-9 which facilitates immunosuppression in cooperation with other immune checkpoint proteins. J Immunother Cancer 2023; 11:jitc-2022-005714. [PMID: 36599470 DOI: 10.1136/jitc-2022-005714] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Galectin-9 is a member of the family of lectin proteins and crucially regulates human immune responses, particularly because of its ability to suppress the anticancer activities of T lymphocytes and natural killer cells. Recent evidence demonstrated that galectin-9 is highly expressed in a wide range of human malignancies including the most aggressive tumors, such as high-grade glioblastomas and pancreatic ductal adenocarcinomas, as well as common malignancies such as breast, lung and colorectal cancers. However, solid tumor cells at rest are known to secrete either very low amounts of galectin-9 or, in most of the cases, do not secrete it at all. Our aims were to elucidate whether T cells can induce galectin-9 secretion in human cancer cells derived from solid malignant tumors and whether this soluble form displays higher systemic immunosuppressive activity compared with the cell surface-based protein. METHODS A wide range of human cancer cell lines derived from solid tumours, keratinocytes and primary embryonic cells were employed, together with helper and cytotoxic T cell lines and human as well as mouse primary T cells. Western blot analysis, ELISA, quantitative reverse transcriptase-PCR, on-cell Western and other measurement techniques were used to conduct the study. Results were validated using in vivo mouse model. RESULTS We discovered that T lymphocytes induce galectin-9 secretion in various types of human cancer cells derived from solid malignant tumors. This was demonstrated to occur via two differential mechanisms: first by translocation of galectin-9 onto the cell surface followed by its proteolytic shedding and second due to autophagy followed by lysosomal secretion. For both mechanisms a protein carrier/trafficker was required, since galectin-9 lacks a secretion sequence. Secreted galectin-9 pre-opsonised T cells and, following interaction with other immune checkpoint proteins, their activity was completely attenuated. As an example, we studied the cooperation of galectin-9 and V-domain Ig-containing suppressor of T cell activation (VISTA) proteins in human cancer cells. CONCLUSION Our results underline a crucial role of galectin-9 in anticancer immune evasion. As such, galectin-9 and regulatory pathways controlling its production should be considered as key targets for immunotherapy in a large number of cancers.
Collapse
Affiliation(s)
- Stephanie Schlichtner
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, Kent, UK
| | - Inna M Yasinska
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, Kent, UK
| | - Gurprit S Lall
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, Kent, UK
| | - Steffen M Berger
- Department of Pediatric Surgery and Department of Biomedical Research, Children's Hospital, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Sabrina Ruggiero
- Department of Pediatric Surgery and Department of Biomedical Research, Children's Hospital, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Dietmar Cholewa
- Department of Pediatric Surgery and Department of Biomedical Research, Children's Hospital, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Nijas Aliu
- Department of Human Genetics, Children's Hospital, Inselspital, University of Bern, Bern, Switzerland
| | - Bernhard F Gibbs
- Department of Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Elizaveta Fasler-Kan
- Department of Pediatric Surgery and Department of Biomedical Research, Children's Hospital, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Vadim V Sumbayev
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, Kent, UK
| |
Collapse
|
16
|
Xiao C, Tian H, Zheng Y, Yang Z, Li S, Fan T, Xu J, Bai G, Liu J, Deng Z, Li C, He J. Glycolysis in tumor microenvironment as a target to improve cancer immunotherapy. Front Cell Dev Biol 2022; 10:1013885. [PMID: 36200045 PMCID: PMC9527271 DOI: 10.3389/fcell.2022.1013885] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/07/2022] [Indexed: 12/03/2022] Open
Abstract
Cancer cells and immune cells all undergo remarkably metabolic reprogramming during the oncogenesis and tumor immunogenic killing processes. The increased dependency on glycolysis is the most typical trait, profoundly involved in the tumor immune microenvironment and cancer immunity regulation. However, how to best utilize glycolytic targets to boost anti-tumor immunity and improve immunotherapies are not fully illustrated. In this review, we describe the glycolytic remodeling of various immune cells within the tumor microenvironment (TME) and the deleterious effects of limited nutrients and acidification derived from enhanced tumor glycolysis on immunological anti-tumor capacity. Moreover, we elucidate the underlying regulatory mechanisms of glycolytic reprogramming, including the crosstalk between metabolic pathways and immune checkpoint signaling. Importantly, we summarize the potential glycolysis-related targets that are expected to improve immunotherapy benefits. Our understanding of metabolic effects on anti-tumor immunity will be instrumental for future therapeutic regimen development.
Collapse
Affiliation(s)
- Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - He Tian
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yujia Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zhenlin Yang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Shuofeng Li
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jiachen Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Guangyu Bai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jingjing Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- *Correspondence: Chunxiang Li, ; Jie He,
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- *Correspondence: Chunxiang Li, ; Jie He,
| |
Collapse
|
17
|
Ouyang X, Gong Y. One Stone, Two Birds: N6-Methyladenosine RNA Modification in Leukemia Stem Cells and the Tumor Immune Microenvironment in Acute Myeloid Leukemia. Front Immunol 2022; 13:912526. [PMID: 35720276 PMCID: PMC9201081 DOI: 10.3389/fimmu.2022.912526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/09/2022] [Indexed: 02/05/2023] Open
Abstract
Acute myeloid leukemia is the most common acute leukemia in adults, with accumulation of abundant blasts and impairment of hematogenic function. Despite great advances in diagnosis and therapy, the overall survival of patients with acute myeloid leukemia remains poor. Leukemia stem cells are the root cause of relapse and chemoresistance in acute myeloid leukemia. The tumor immune microenvironment is another trigger to induce recurrence and drug resistance. Understanding the underlying factors influencing leukemia stem cells and the tumor immune microenvironment is an urgent and unmet need. Intriguingly, N6-methyladenosine, the most widespread internal mRNA modification in eukaryotes, is found to regulate both leukemia stem cells and the tumor immune microenvironment. Methyltransferases and demethylases cooperatively make N6-methyladenosine modification reversible and dynamic. Increasing evidence demonstrates that N6-methyladenosine modification extensively participates in tumorigenesis and progression in various cancers, including acute myeloid leukemia. In this review, we summarize the current progress in studies on the functions of N6-methyladenosine modification in acute myeloid leukemia, especially in leukemia stem cells and the tumor immune microenvironment. We generalize the landscape of N6-methyladenosine modification in self-renewal of leukemia stem cells and immune microenvironment regulation, as well as in the initiation, growth, proliferation, differentiation, and apoptosis of leukemia cells. In addition, we further explore the clinical application of N6-methyladenosine modification in diagnosis, prognostic stratification, and effect evaluation. Considering the roles of N6-methyladenosine modification in leukemia stem cells and the tumor immune microenvironment, we propose targeting N6-methyladenosine regulators as one stone to kill two birds for acute myeloid leukemia treatment.
Collapse
Affiliation(s)
- Xianfeng Ouyang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China.,Department of Hematology, Jiujiang First People's Hospital, Jiujiang, China
| | - Yuping Gong
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Schlichtner S, Yasinska IM, Ruggiero S, Berger SM, Aliu N, Prunk M, Kos J, Meyer NH, Gibbs BF, Fasler-Kan E, Sumbayev VV. Expression of the Immune Checkpoint Protein VISTA Is Differentially Regulated by the TGF-β1 - Smad3 Signaling Pathway in Rapidly Proliferating Human Cells and T Lymphocytes. Front Med (Lausanne) 2022; 9:790995. [PMID: 35223897 PMCID: PMC8866318 DOI: 10.3389/fmed.2022.790995] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 01/17/2022] [Indexed: 01/25/2023] Open
Abstract
Immune checkpoint proteins play crucial roles in human embryonic development but are also used by cancer cells to escape immune surveillance. These proteins and biochemical pathways associated with them form a complex machinery capable of blocking the ability of cytotoxic immune lymphoid cells to attack cancer cells and, ultimately, to fully suppress anti-tumor immunity. One of the more recently discovered immune checkpoint proteins is V-domain Ig-containing suppressor of T cell activation (VISTA), which plays a crucial role in anti-cancer immune evasion pathways. The biochemical mechanisms underlying regulation of VISTA expression remain unknown. Here, we report for the first time that VISTA expression is controlled by the transforming growth factor beta type 1 (TGF-β)-Smad3 signaling pathway. However, in T lymphocytes, we found that VISTA expression was differentially regulated by TGF-β depending on their immune profile. Taken together, our results demonstrate the differential biochemical control of VISTA expression in human T cells and various types of rapidly proliferating cells, including cancer cells, fetal cells and keratinocytes.
Collapse
Affiliation(s)
- Stephanie Schlichtner
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - Inna M Yasinska
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - Sabrina Ruggiero
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Steffen M Berger
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Nijas Aliu
- Department of Human Genetics, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Mateja Prunk
- Department of Biotechnology, JoŽef Stefan Institute, Ljubljana, Slovenia
| | - Janko Kos
- Department of Biotechnology, JoŽef Stefan Institute, Ljubljana, Slovenia.,Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - N Helge Meyer
- Division of Experimental Allergology and Immunodermatology, Department of Human Medicine, University of Oldenburg, Oldenburg, Germany.,Division of General and Visceral Surgery, Department of Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Bernhard F Gibbs
- Division of Experimental Allergology and Immunodermatology, Department of Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Elizaveta Fasler-Kan
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern, Bern, Switzerland.,Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Vadim V Sumbayev
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| |
Collapse
|
19
|
Rezaei M, Tan J, Zeng C, Li Y, Ganjalikhani-Hakemi M. TIM-3 in Leukemia; Immune Response and Beyond. Front Oncol 2021; 11:753677. [PMID: 34660319 PMCID: PMC8514831 DOI: 10.3389/fonc.2021.753677] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/10/2021] [Indexed: 02/05/2023] Open
Abstract
T cell immunoglobulin and mucin domain 3 (TIM-3) expression on malignant cells has been reported in some leukemias. In myelodysplastic syndrome (MDS), increased TIM-3 expression on TH1 cells, regulatory T cells, CD8+ T cells, and hematopoietic stem cells (HSCs), which play a role in the proliferation of blasts and induction of immune escape, has been reported. In AML, several studies have reported overexpression of TIM-3 on leukemia stem cells (LSCs) but not on healthy HSCs. Overexpression of TIM-3 on exhausted CD4+ and CD8+ T cells and leukemic cells in CML, ALL, and CLL patients could be a prognostic risk factor for poor therapeutic response and relapse in patients. Currently, several TIM-3 inhibitors are used in clinical trials for leukemias, and some have shown encouraging response rates for MDS and AML treatment. For AML immunotherapy, blockade TIM-3 may have dual effects: directly inhibiting AML cell proliferation and restoring T cell function. However, blockade of PD-1 and TIM-3 fails to restore the function of exhausted CD8+ T cells in the early clinical stages of CLL, indicating that the effects of TIM-3 blockade may be different in AML and other leukemias. Thus, further studies are required to evaluate the efficacy of TIM-3 inhibitors in different types and stages of leukemia. In this review, we summarize the biological functions of TIM-3 and its contribution as it relates to leukemias. We also discuss the effects of TIM-3 blockade in hematological malignancies and clinical trials of TIM-3 for leukemia therapy.
Collapse
Affiliation(s)
- Mahnaz Rezaei
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Jiaxiong Tan
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Chengwu Zeng
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Yangqiu Li
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Mazdak Ganjalikhani-Hakemi
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Acquired Immunodeficiency Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
20
|
Schlichtner S, Meyer NH, Yasinska IM, Aliu N, Berger SM, Gibbs BF, Fasler-Kan E, Sumbayev VV. Functional role of galectin-9 in directing human innate immune reactions to Gram-negative bacteria and T cell apoptosis. Int Immunopharmacol 2021; 100:108155. [PMID: 34543981 DOI: 10.1016/j.intimp.2021.108155] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 12/26/2022]
Abstract
Galectin-9 is a member of the galectin family of proteins, which were first identified to specifically bind to carbohydrates containing β-galactosides. Galectin-9 is conserved through evolution and recent evidence demonstrated its involvement in innate immune reactions to bacterial infections as well as the suppression of cytotoxic immune responses of T and natural killer cells. However, the molecular mechanisms underlying such differential immunological functions of galectin-9 remain largely unknown. In this work we confirmed that soluble galectin-9 derived from macrophages binds to Gram-negative bacteria by interacting with lipopolysaccharide (LPS), which forms their cell wall. This opsonisation effect most likely interferes with the mobility of bacteria leading to their phagocytosis by innate immune cells. Galectin-9-dependent opsonisation also promotes the innate immune reactions of macrophages to these bacteria and significantly enhances the production of pro-inflammatory cytokines - interleukin (IL) 6, IL-1β and tumour necrosis factor alpha (TNF-α). In contrast, galectin-9 did not bind peptidoglycan (PGN), which forms the cell wall of Gram-positive bacteria. Moreover, galectin-9 associated with cellular surfaces (studied in primary human embryonic cells) was not involved in the interaction with bacteria or bacterial colonisation. However, galectin-9 expressed on the surface of primary human embryonic cells, as well as soluble forms of galectin-9, were able to target T lymphocytes and caused apoptosis in T cells expressing granzyme B. Furthermore, "opsonisation" of T cells by galectin-9 led to the translocation of phosphatidylserine onto the cell surface and subsequent phagocytosis by macrophages through Tim-3, the receptor, which recognises both galectin-9 and phosphatidylserine as ligands.
Collapse
Affiliation(s)
- Stephanie Schlichtner
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - N Helge Meyer
- Division of Experimental Allergology and Immunodermatology, Department of Human Medicine, University of Oldenburg, Oldenburg, Germany; Division of General and Visceral Surgery, Department of Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Inna M Yasinska
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - Nijas Aliu
- Department of Human Genetics, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Steffen M Berger
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Bernhard F Gibbs
- Division of Experimental Allergology and Immunodermatology, Department of Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Elizaveta Fasler-Kan
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern, Bern, Switzerland; Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland.
| | - Vadim V Sumbayev
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom.
| |
Collapse
|
21
|
Kikushige Y. TIM-3 in normal and malignant hematopoiesis: Structure, function, and signaling pathways. Cancer Sci 2021; 112:3419-3426. [PMID: 34159709 PMCID: PMC8409405 DOI: 10.1111/cas.15042] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/03/2021] [Accepted: 06/20/2021] [Indexed: 12/15/2022] Open
Abstract
Acute myeloid leukemia (AML) is hierarchically organized by self-renewing leukemic stem cells (LSCs). LSCs originate from hematopoietic stem cells (HSCs) by acquiring multistep leukemogenic events. To specifically eradicate LSCs, while keeping normal HSCs intact, the discrimination of LSCs from HSCs is important. We have identified T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) as an LSC-specific surface molecule in human myeloid malignancies and demonstrated its essential function in maintaining the self-renewal ability of LSCs. TIM-3 has been intensively investigated as a "coinhibitory" or "immune checkpoint" molecule of T cells. However, little is known about its distinct function in T cells and myeloid malignancies. In this review, we discuss the structure of TIM-3 and its function in normal blood cells and LSCs, emphasizing the specific signaling pathways involved, as well as the therapeutic applications of TIM-3 molecules in human myeloid malignancies.
Collapse
Affiliation(s)
- Yoshikane Kikushige
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, Japan.,Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| |
Collapse
|
22
|
Teo Hansen Selnø A, Schlichtner S, Yasinska IM, Sakhnevych SS, Fiedler W, Wellbrock J, Berger SM, Klenova E, Gibbs BF, Fasler-Kan E, Sumbayev VV. High Mobility Group Box 1 (HMGB1) Induces Toll-Like Receptor 4-Mediated Production of the Immunosuppressive Protein Galectin-9 in Human Cancer Cells. Front Immunol 2021; 12:675731. [PMID: 34234778 PMCID: PMC8255966 DOI: 10.3389/fimmu.2021.675731] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/27/2021] [Indexed: 01/19/2023] Open
Abstract
High mobility group box 1 (HMGB1) is a non-histone protein which is predominantly localised in the cell nucleus. However, stressed, dying, injured or dead cells can release this protein into the extracellular matrix passively. In addition, HMGB1 release was observed in cancer and immune cells where this process can be triggered by various endogenous as well as exogenous stimuli. Importantly, released HMGB1 acts as a so-called "danger signal" and could impact on the ability of cancer cells to escape host immune surveillance. However, the molecular mechanisms underlying the functional role of HMGB1 in determining the capability of human cancer cells to evade immune attack remain unclear. Here we report that the involvement of HMGB1 in anti-cancer immune evasion is determined by Toll-like receptor (TLR) 4, which recognises HMGB1 as a ligand. We found that HGMB1 induces TLR4-mediated production of transforming growth factor beta type 1 (TGF-β), displaying autocrine/paracrine activities. TGF-β induces production of the immunosuppressive protein galectin-9 in cancer cells. In TLR4-positive cancer cells, HMGB1 triggers the formation of an autocrine loop which induces galectin-9 expression. In malignant cells lacking TLR4, the same effect could be triggered by HMGB1 indirectly through TLR4-expressing myeloid cells present in the tumour microenvironment (e. g. tumour-associated macrophages).
Collapse
Affiliation(s)
- Anette Teo Hansen Selnø
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - Stephanie Schlichtner
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - Inna M Yasinska
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - Svetlana S Sakhnevych
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - Walter Fiedler
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jasmin Wellbrock
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Steffen M Berger
- Department of Pediatric Surgery, Department of Biomedical Research, Children's Hospital, Inselspital, University of Bern, Bern, Switzerland.,Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Elena Klenova
- School of Biological Sciences, University of Essex, Colchester, United Kingdom
| | - Bernhard F Gibbs
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom.,Division of Experimental Allergy and Immunodermatology, University of Oldenburg, Oldenburg, Germany
| | - Elizaveta Fasler-Kan
- Department of Pediatric Surgery, Department of Biomedical Research, Children's Hospital, Inselspital, University of Bern, Bern, Switzerland.,Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Vadim V Sumbayev
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| |
Collapse
|
23
|
Zhu C, Dixon KO, Newcomer K, Gu G, Xiao S, Zaghouani S, Schramm MA, Wang C, Zhang H, Goto K, Christian E, Rangachari M, Rosenblatt-Rosen O, Okada H, Mak T, Singer M, Regev A, Kuchroo V. Tim-3 adaptor protein Bat3 is a molecular checkpoint of T cell terminal differentiation and exhaustion. SCIENCE ADVANCES 2021; 7:eabd2710. [PMID: 33931442 PMCID: PMC8087420 DOI: 10.1126/sciadv.abd2710] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 03/10/2021] [Indexed: 05/03/2023]
Abstract
T cell exhaustion has been associated with poor prognosis in persistent viral infection and cancer. Conversely, in the context of autoimmunity, T cell exhaustion has been favorably correlated with long-term clinical outcome. Understanding the development of exhaustion in autoimmune settings may provide underlying principles that can be exploited to quell autoreactive T cells. Here, we demonstrate that the adaptor molecule Bat3 acts as a molecular checkpoint of T cell exhaustion, with deficiency of Bat3 promoting a profound exhaustion phenotype, suppressing autoreactive T cell-mediated neuroinflammation. Mechanistically, Bat3 acts as a critical mTORC2 inhibitor to suppress Akt function. As a result, Bat3 deficiency leads to increased Akt activity and FoxO1 phosphorylation, indirectly promoting Prdm1 expression. Transcriptional analysis of Bat3 -/- T cells revealed up-regulation of dysfunction-associated genes, concomitant with down-regulation of genes associated with T cell effector function, suggesting that absence of Bat3 can trigger T cell dysfunction even under highly proinflammatory autoimmune conditions.
Collapse
Affiliation(s)
- Chen Zhu
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Karen O Dixon
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kathleen Newcomer
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
- Department of Data Sciences, DFCI, Boston, MA 02215, USA
| | - Guangxiang Gu
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Sheng Xiao
- Celsius Therapeutics, Cambridge, MA 02139, USA
| | - Sarah Zaghouani
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Markus A Schramm
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Chao Wang
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Huiyuan Zhang
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Kouichiro Goto
- The Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, University of Health Network, Toronto, Ontario M5G 2M9, Canada
| | | | - Manu Rangachari
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, QC, Québec, Canada
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, QC, Canada
| | | | - Hitoshi Okada
- Department of Biochemistry, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Tak Mak
- The Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, University of Health Network, Toronto, Ontario M5G 2M9, Canada
| | - Meromit Singer
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
- Department of Data Sciences, DFCI, Boston, MA 02215, USA
| | - Aviv Regev
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Biology, Koch Institute and Ludwig Center, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Vijay Kuchroo
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
24
|
Wang Z, Chen J, Wang M, Zhang L, Yu L. One Stone, Two Birds: The Roles of Tim-3 in Acute Myeloid Leukemia. Front Immunol 2021; 12:618710. [PMID: 33868234 PMCID: PMC8047468 DOI: 10.3389/fimmu.2021.618710] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 03/18/2021] [Indexed: 12/11/2022] Open
Abstract
T cell immunoglobulin and mucin protein 3 (Tim-3) is an immune checkpoint and plays a vital role in immune responses during acute myeloid leukemia (AML). Targeting Tim-3 kills two birds with one stone by balancing the immune system and eliminating leukemia stem cells (LSCs) in AML. These functions make Tim-3 a potential target for curing AML. This review mainly discusses the roles of Tim-3 in the immune system in AML and as an AML LSC marker, which sheds new light on the role of Tim-3 in AML immunotherapy.
Collapse
Affiliation(s)
- Zhiding Wang
- Department of Hematology and Oncology, International Cancer Center, Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University Health Science Center, Shenzhen, China.,Beijing Institute of Basic Medical Sciences, Beijing, China.,Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing, China
| | - Jinghong Chen
- Department of Hematology and Oncology, International Cancer Center, Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University Health Science Center, Shenzhen, China
| | - Mengzhen Wang
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing, China
| | - Linlin Zhang
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing, China
| | - Li Yu
- Department of Hematology and Oncology, International Cancer Center, Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University Health Science Center, Shenzhen, China.,Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
25
|
Transforming growth factor beta type 1 (TGF-β) and hypoxia-inducible factor 1 (HIF-1) transcription complex as master regulators of the immunosuppressive protein galectin-9 expression in human cancer and embryonic cells. Aging (Albany NY) 2020; 12:23478-23496. [PMID: 33295886 PMCID: PMC7762483 DOI: 10.18632/aging.202343] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/15/2020] [Indexed: 12/11/2022]
Abstract
Galectin-9 is one of the key proteins employed by a variety of human malignancies to suppress anti-cancer activities of cytotoxic lymphoid cells and thus escape immune surveillance. Human cancer cells in most cases express higher levels of galectin-9 compared to non-transformed cells. However, the biochemical mechanisms underlying this phenomenon remain unclear. Here we report for the first time that in human cancer as well as embryonic cells, the transcription factors hypoxia-inducible factor 1 (HIF-1) and activator protein 1 (AP-1) are involved in upregulation of transforming growth factor beta 1 (TGF-β1) expression, leading to activation of the transcription factor Smad3 through autocrine action. This process triggers upregulation of galectin-9 expression in both malignant (mainly in breast and colorectal cancer as well as acute myeloid leukaemia (AML)) and embryonic cells. The effect, however, was not observed in mature non-transformed human cells. TGF-β1-activated Smad3 therefore displays differential behaviour in human cancer and embryonic vs non-malignant cells. This study uncovered a self-supporting biochemical mechanism underlying high levels of galectin-9 expression operated by the human cancer and embryonic cells employed in our investigations. Our results suggest the possibility of using the TGF-β1 signalling pathway as a potential highly efficient target for cancer immunotherapy.
Collapse
|
26
|
Ganjalikhani Hakemi M, Jafarinia M, Azizi M, Rezaeepoor M, Isayev O, Bazhin AV. The Role of TIM-3 in Hepatocellular Carcinoma: A Promising Target for Immunotherapy? Front Oncol 2020; 10:601661. [PMID: 33425759 PMCID: PMC7793963 DOI: 10.3389/fonc.2020.601661] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/26/2020] [Indexed: 02/05/2023] Open
Abstract
One of the most common tumors in the world is hepatocellular carcinoma (HCC), and its mortality rates are still on the rise, so addressing it is considered an important challenge for universal health. Despite the various treatments that have been developed over the past decades, the prognosis for advanced liver cancer is still poor. Recently, tumor immunotherapy has opened new opportunities for suppression of tumor progression, recurrence, and metastasis. Besides this, investigation into this malignancy due to high immune checkpoint expression and the change of immunometabolic programming in immune cells and tumor cells is highly considered. Because anti-cytotoxic T lymphocyte-associated protein (CTLA)-4 antibodies and anti-programmed cell death protein (PD)-1 antibodies have shown therapeutic effects in various cancers, studies have shown that T cell immunoglobulin mucin-3 (TIM-3), a new immune checkpoint molecule, plays an important role in the development of HCC. In this review, we summarize the recent findings on signal transduction events of TIM-3, its role as a checkpoint target for HCC therapy, and the immunometabolic situation in the progression of HCC.
Collapse
Affiliation(s)
| | - Morteza Jafarinia
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahdieh Azizi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahsa Rezaeepoor
- Department of Immunology, School of Medicine, Hamedan University of Medical Sciences, Hamedan, Iran
| | - Orkhan Isayev
- Department of Cytology, Embryology and Histology, Azerbaijan Medical University, Baku, Azerbaijan
- Genetic Resources Institute, Azerbaijan National Academy of Scince, Baku, Azerbaijan
| | - Alexandr V. Bazhin
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| |
Collapse
|
27
|
Yasinska IM, Meyer NH, Schlichtner S, Hussain R, Siligardi G, Casely-Hayford M, Fiedler W, Wellbrock J, Desmet C, Calzolai L, Varani L, Berger SM, Raap U, Gibbs BF, Fasler-Kan E, Sumbayev VV. Ligand-Receptor Interactions of Galectin-9 and VISTA Suppress Human T Lymphocyte Cytotoxic Activity. Front Immunol 2020; 11:580557. [PMID: 33329552 PMCID: PMC7715031 DOI: 10.3389/fimmu.2020.580557] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/23/2020] [Indexed: 12/25/2022] Open
Abstract
Acute myeloid leukemia (AML), a blood/bone marrow cancer, is a severe and often fatal malignancy. AML cells are capable of impairing the anti-cancer activities of cytotoxic lymphoid cells. This includes the inactivation of natural killer (NK) cells and killing of T lymphocytes. Here we report for the first time that V-domain Ig-containing suppressor of T cell activation (VISTA), a protein expressed by T cells, recognizes galectin-9 secreted by AML cells as a ligand. Importantly, we found that soluble VISTA released by AML cells enhances the effect of galectin-9, most likely by forming multiprotein complexes on the surface of T cells and possibly creating a molecular barrier. These events cause changes in the plasma membrane potential of T cells leading to activation of granzyme B inside cytotoxic T cells, resulting in apoptosis.
Collapse
Affiliation(s)
- Inna M Yasinska
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - N Helge Meyer
- Division of Experimental Allergology and Immunodermatology, University of Oldenburg, Oldenburg, Germany
| | - Stephanie Schlichtner
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | | | | | - Maxwell Casely-Hayford
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - Walter Fiedler
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jasmin Wellbrock
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cloe Desmet
- European Commission Joint Research Centre, Ispra, Italy
| | | | - Luca Varani
- Institute for Research in Biomedicine, Universita' della Svizzera Italiana (USI), Bellinzona, Switzerland
| | - Steffen M Berger
- Department of Pediatric Surgery, Department of Biomedical Research, Children's Hospital, Inselspital, University of Bern, Bern, Switzerland
| | - Ulrike Raap
- Division of Experimental Allergology and Immunodermatology, University of Oldenburg, Oldenburg, Germany
| | - Bernhard F Gibbs
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom.,Division of Experimental Allergology and Immunodermatology, University of Oldenburg, Oldenburg, Germany
| | - Elizaveta Fasler-Kan
- Department of Pediatric Surgery, Department of Biomedical Research, Children's Hospital, Inselspital, University of Bern, Bern, Switzerland.,Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Vadim V Sumbayev
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| |
Collapse
|
28
|
Zhang D, Jiang F, Zaynagetdinov R, Huang H, Sood VD, Wang H, Zhao X, Jenkins MH, Ji Q, Wang Y, Nannemann DP, Musil D, Wesolowski J, Paoletti A, Bartholomew T, Derner MG, An Q, Iffland C, Halle JP. Identification and characterization of M6903, an antagonistic anti-TIM-3 monoclonal antibody. Oncoimmunology 2020; 9:1744921. [PMID: 32313722 PMCID: PMC7153820 DOI: 10.1080/2162402x.2020.1744921] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 02/04/2020] [Accepted: 02/25/2020] [Indexed: 02/06/2023] Open
Abstract
T cell immunoglobulin and mucin domain-3 (TIM-3) is an immune checkpoint that regulates normal immune responses but can be exploited by tumor cells to evade immune surveillance. TIM-3 is primarily expressed on immune cells, particularly on dysfunctional and exhausted T cells, and engagement of TIM-3 with its ligands promotes TIM-3-mediated T cell inhibition. Antagonistic ligand-blocking anti-TIM-3 antibodies have the potential to abrogate T cell inhibition, activate antigen-specific T cells, and enhance anti-tumor immunity. Here we describe M6903, a fully human anti-TIM-3 antibody without effector function and with high affinity and selectivity to TIM-3. We demonstrate that M6903 blocks the binding of TIM-3 to three of its ligands, phosphatidylserine (PtdSer), carcinoembryonic antigen cell adhesion-related molecule 1 (CEACAM1), and galectin 9 (Gal-9). These results are supported by an atomic resolution crystal structure and functional assays, which demonstrate that M6903 monotherapy enhanced T cell activation. This activation was further enhanced by the combination of M6903 with bintrafusp alfa, a bifunctional fusion protein that simultaneously blocks the transforming growth factor-β (TGF-β) and programmed death ligand 1 (PD-L1) pathways. M6903 and bintrafusp alfa combination therapy also enhanced anti-tumor efficacy in huTIM-3 knock-in mice, relative to either monotherapy. These in vitro and in vivo data, along with favorable pharmacokinetics in marmoset monkeys, suggest that M6903 as a monotherapy warrants further pre-clinical assessment and that M6903 and bintrafusp alfa may be a promising combination therapy in the clinic.
Collapse
Affiliation(s)
- Dong Zhang
- Department of Immuno-Oncology, EMD Serono Research and Development Institute, Billerica, MA, USA
| | - Feng Jiang
- Department of Immuno-Oncology, EMD Serono Research and Development Institute, Billerica, MA, USA
| | - Rinat Zaynagetdinov
- Department of Immuno-Oncology, EMD Serono Research and Development Institute, Billerica, MA, USA
| | - Hui Huang
- Department of Immuno-Oncology, EMD Serono Research and Development Institute, Billerica, MA, USA
| | - Vanita D Sood
- Discovery and Development Technologies, EMD Serono Research and Development Institute, Billerica, MA, USA
| | - Hong Wang
- Department of Immuno-Oncology, EMD Serono Research and Development Institute, Billerica, MA, USA
| | - Xinyan Zhao
- Discovery and Development Technologies, EMD Serono Research and Development Institute, Billerica, MA, USA
| | - Molly H Jenkins
- Department of Immuno-Oncology, EMD Serono Research and Development Institute, Billerica, MA, USA
| | - Qingyong Ji
- Discovery and Development Technologies, EMD Serono Research and Development Institute, Billerica, MA, USA
| | - Youbin Wang
- Discovery and Development Technologies, EMD Serono Research and Development Institute, Billerica, MA, USA
| | - David P Nannemann
- Discovery and Development Technologies, EMD Serono Research and Development Institute, Billerica, MA, USA
| | - Djordje Musil
- Discovery and Development Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - John Wesolowski
- Discovery and Development Technologies, EMD Serono Research and Development Institute, Billerica, MA, USA
| | - Andrea Paoletti
- Discovery and Development Technologies, Merck Healthcare KGaA, Colleretto Giacosa, Italy
| | - Tin Bartholomew
- Department of Immuno-Oncology, EMD Serono Research and Development Institute, Billerica, MA, USA
| | - Melissa G Derner
- Department of Immuno-Oncology, EMD Serono Research and Development Institute, Billerica, MA, USA
| | - Qi An
- Discovery and Development Technologies, EMD Serono Research and Development Institute, Billerica, MA, USA
| | - Christel Iffland
- Discovery and Development Technologies, EMD Serono Research and Development Institute, Billerica, MA, USA
| | - Joern-Peter Halle
- Department of Immuno-Oncology, EMD Serono Research and Development Institute, Billerica, MA, USA.,Department of Immuno-Oncology, Merck Healthcare KGaA, Darmstadt, Germany
| |
Collapse
|
29
|
Huang R, Meng T, Zhu R, Zhao L, Song D, Yin H, Huang Z, Cheng L, Zhang J. The Integrated Transcriptome Bioinformatics Analysis Identifies Key Genes and Cellular Components for Spinal Cord Injury-Related Neuropathic Pain. Front Bioeng Biotechnol 2020; 8:101. [PMID: 32140464 PMCID: PMC7042182 DOI: 10.3389/fbioe.2020.00101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/03/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is one of the most devastating diseases with a high incidence rate around the world. SCI-related neuropathic pain (NeP) is a common complication, whereas its pathomechanism is still unclear. The purpose of this study is to identify key genes and cellular components for SCI-related NeP by an integrated transcriptome bioinformatics analysis. METHODS The gene expression profile of 25 peripheral blood samples from chronic phase SCI patients (E-GEOD-69901) and 337 normal peripheral blood samples were downloaded from ArrayExpress and Genotype-Tissue Expression Portal (GTEx), respectively. A total of 3,368 normal peripheral blood mononuclear cells (PBMC) were download from Sequence Read Archive (SRA713577). Non-parametric tests were used to evaluate the association between all of differential expression genes (DEGs) and SCI-related NeP. CellPhoneDB algorithm was performed to identify the ligand-receptor interactions and their cellular localization among single PBMCs. Transcription factor (TF) enrichment analysis and Gene Set Variation Analysis (GSVA) were used to identify the potential upstream regulatory TFs and downstream signaling pathways, respectively. Co-expression analysis among significantly enriched TFs, key cellular communication genes and differentially expressed signaling pathways were performed to identify key genes and cellular components for SCI-related NeP. RESULTS A total of 2,314 genes were identified as DEGs between the experimental and the control group. Five proteins (ADRB2, LGALS9, PECAM1, HAVCR2, LRP1) were identified in the overlap of proteins in the significant ligand-receptor interactions of PBMCs and protein-protein interaction (PPI) network based on the DEGs. Only HAVCR2 was significantly associated with NeP (P = 0.005). Besides, the co-expression analysis revealed that TF YY1 had significantly co-expression pattern with cellular communication receptor HAVCR2 (R = -0.54, P < 0.001) in NK cells while HAVCR2 was also co-expressed with mTOR signaling pathway (R = 0.57, P < 0.001). The results of RT-qPCR and external dataset validation supported the signaling axis with the most significant co-expression patterns. CONCLUSION In peripheral blood of chronic SCI, HAVCR2 might act as a key receptor on the surface of NK cells and interact with ligand LGALS9 secreted by CD14+ monocytes, inhibiting NK cells through mTOR signaling pathway and ultimately predicting the occurrence of SCI-related NeP. This hypothetical signaling axis may provide prognostic biomarkers and therapeutic targets for SCI-related NeP.
Collapse
Affiliation(s)
- Runzhi Huang
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tong Meng
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Rui Zhu
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lijuan Zhao
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dianwen Song
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huabin Yin
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zongqiang Huang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liming Cheng
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jie Zhang
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Prevention, Tongji University School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
30
|
Zhang Y, Zheng J. Functions of Immune Checkpoint Molecules Beyond Immune Evasion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1248:201-226. [PMID: 32185712 DOI: 10.1007/978-981-15-3266-5_9] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Immune checkpoint molecules, including inhibitory and stimulatory immune checkpoint molecules, are defined as ligand-receptor pairs that exert inhibitory or stimulatory effects on immune responses. Most of the immune checkpoint molecules that have been described so far are expressed on cells of the adaptive immune system, particularly on T cells, and of the innate immune system. They are crucial for maintaining the self-tolerance and modulating the length and magnitude of immune responses of effectors in different tissues to minimize the tissue damage. More and more evidences have shown that inhibitory or stimulatory immune checkpoint molecules are expressed on a sizeable fraction of tumor types. Although the main function of tumor cell-associated immune checkpoint molecules is considered to mediate the immune evasion, it has been reported that the immune checkpoint molecules expressed on tumor cells also play important roles in the maintenance of many malignant behaviors, including self-renewal, epithelial-mesenchymal transition, metastasis, drug resistance, anti-apoptosis, angiogenesis, or enhanced energy metabolisms. In this section, we mainly focus on delineating the roles of the tumor cell-associated immune checkpoint molecules beyond immune evasion, such as PD-L1, PD-1, B7-H3, B7-H4, LILRB1, LILRB2, TIM3, CD47, CD137, and CD70.
Collapse
Affiliation(s)
- Yaping Zhang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Junke Zheng
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
31
|
Yasinska IM, Sakhnevych SS, Pavlova L, Teo Hansen Selnø A, Teuscher Abeleira AM, Benlaouer O, Gonçalves Silva I, Mosimann M, Varani L, Bardelli M, Hussain R, Siligardi G, Cholewa D, Berger SM, Gibbs BF, Ushkaryov YA, Fasler-Kan E, Klenova E, Sumbayev VV. The Tim-3-Galectin-9 Pathway and Its Regulatory Mechanisms in Human Breast Cancer. Front Immunol 2019; 10:1594. [PMID: 31354733 PMCID: PMC6637653 DOI: 10.3389/fimmu.2019.01594] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 06/26/2019] [Indexed: 01/09/2023] Open
Abstract
Human cancer cells operate a variety of effective molecular and signaling mechanisms which allow them to escape host immune surveillance and thus progress the disease. We have recently reported that the immune receptor Tim-3 and its natural ligand galectin-9 are involved in the immune escape of human acute myeloid leukemia (AML) cells. These cells use the neuronal receptor latrophilin 1 (LPHN1) and its ligand fibronectin leucine rich transmembrane protein 3 (FLRT3, and possibly other ligands) to trigger the pathway. We hypothesized that the Tim-3-galectin-9 pathway may be involved in the immune escape of cancer cells of different origins. We found that studied breast tumors expressed significantly higher levels of both galectin-9 and Tim-3 compared to healthy breast tissues of the same patients and that these proteins were co-localized. Increased levels of LPHN2 and expressions of LPHN3 as well as FLRT3 were also detected in breast tumor cells. Activation of this pathway facilitated the translocation of galectin-9 onto the tumor cell surface, however no secretion of galectin-9 by tumor cells was observed. Surface-based galectin-9 was able to protect breast carcinoma cells against cytotoxic T cell-induced death. Furthermore, we found that cell lines from brain, colorectal, kidney, blood/mast cell, liver, prostate, lung, and skin cancers expressed detectable amounts of both Tim-3 and galectin-9 proteins. The majority of cell lines expressed one of the LPHN isoforms and FLRT3. We conclude that the Tim-3-galectin-9 pathway is operated by a wide range of human cancer cells and is possibly involved in prevention of anti-tumor immunity.
Collapse
Affiliation(s)
- Inna M Yasinska
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - Svetlana S Sakhnevych
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - Ludmila Pavlova
- School of Biological Sciences, University of Essex, Colchester, United Kingdom
| | - Anette Teo Hansen Selnø
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - Ana Maria Teuscher Abeleira
- Department of Pediatric Surgery, Department of Biomedical Research, Children's Hospital, Inselspital, University of Bern, Bern, Switzerland.,Zentrum Für Medizinische Bildung, Biomedizinische Analytik HF, Bern, Switzerland
| | - Ouafa Benlaouer
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - Isabel Gonçalves Silva
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - Marianne Mosimann
- Department of Pediatric Surgery, Department of Biomedical Research, Children's Hospital, Inselspital, University of Bern, Bern, Switzerland.,Zentrum Für Medizinische Bildung, Biomedizinische Analytik HF, Bern, Switzerland
| | - Luca Varani
- Institute for Research in Biomedicine, Universita' della Svizzera italiana, Bellinzona, Switzerland
| | - Marco Bardelli
- Institute for Research in Biomedicine, Universita' della Svizzera italiana, Bellinzona, Switzerland
| | | | | | - Dietmar Cholewa
- Department of Pediatric Surgery, Department of Biomedical Research, Children's Hospital, Inselspital, University of Bern, Bern, Switzerland
| | - Steffen M Berger
- Department of Pediatric Surgery, Department of Biomedical Research, Children's Hospital, Inselspital, University of Bern, Bern, Switzerland
| | - Bernhard F Gibbs
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom.,Division of Experimental Allergology and Immunodermatology, University of Oldenburg, Oldenburg, Germany
| | - Yuri A Ushkaryov
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - Elizaveta Fasler-Kan
- Department of Pediatric Surgery, Department of Biomedical Research, Children's Hospital, Inselspital, University of Bern, Bern, Switzerland.,Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Elena Klenova
- School of Biological Sciences, University of Essex, Colchester, United Kingdom
| | - Vadim V Sumbayev
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| |
Collapse
|
32
|
Wang H, Kaur G, Sankin AI, Chen F, Guan F, Zang X. Immune checkpoint blockade and CAR-T cell therapy in hematologic malignancies. J Hematol Oncol 2019; 12:59. [PMID: 31186046 PMCID: PMC6558778 DOI: 10.1186/s13045-019-0746-1] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 05/27/2019] [Indexed: 12/27/2022] Open
Abstract
Harnessing the power of the immune system to recognize and eliminate cancer cells is a longtime exploration. In the past decade, monoclonal antibody (mAb)-based immune checkpoint blockade (ICB) and chimeric antigen receptor T (CAR-T) cell therapy have proven to be safe and effective in hematologic malignancies. Despite the unprecedented success of ICB and CAR-T therapy, only a subset of patients can benefit partially due to immune dysfunction and lack of appropriate targets. Here, we review the preclinical and clinical advances of CTLA-4 and PD-L1/PD-1-based ICB and CD19-specific CAR-T cell therapy in hematologic malignancies. We also discuss the basic research and ongoing clinical trials on emerging immune checkpoints (Galectin-9/Tim-3, CD70/CD27, LAG-3, and LILRBs) and on new targets for CAR-T cell therapy (CD22, CD33, CD123, BCMA, CD38, and CD138) for the treatment of hematologic malignancies.
Collapse
Affiliation(s)
- Hao Wang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Gurbakhash Kaur
- Department of Medical Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Alexander I Sankin
- Department of Urology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Fuxiang Chen
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Fangxia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Department of Medical Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Department of Urology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
33
|
Zheng Y, Feng W, Wang YJ, Sun Y, Shi G, Yu Q. Galectins as potential emerging key targets in different types of leukemia. Eur J Pharmacol 2018; 844:73-78. [PMID: 30452910 DOI: 10.1016/j.ejphar.2018.11.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/20/2018] [Accepted: 11/14/2018] [Indexed: 01/11/2023]
Abstract
Galectins are carbohydrate-binding proteins and these have very high affinity for β-galactoside containing glycoproteins and glycolipids. Amongst sixteen types of galectin, the role of galectin 1, 3, 9 and 12 is defined in the development and progression of different types of leukemia including acute myeloid leukemia, acute promyelocytic leukemia, B-cell precursor acute lymphoblastic leukemia, adult T cell leukemia and chronic lymphocytic leukemia. There are multiple mechanisms through which these galectins may affect tumor proliferation. These may include increased production of tumor resistance conferring proteins such as multidrug resistance (MDR-1) and myeloid cell leukemia (MCL-1). Moreover, galectin-9 may act on Tim-3 receptors present on the circulating CD8+ T cells to impair immune system function and the latter provide an ideal environment for the proliferation of leukemic cells. The present review describes the role and mechanisms involved in galectin-mediated development and progression of different types of leukemia.
Collapse
Affiliation(s)
- Yan Zheng
- The Department of Anesthesia, China-Japan Union Hospital of Jilin University, China.
| | - Wei Feng
- The Department of Anesthesia, China-Japan Union Hospital of Jilin University, China
| | - Yu-Juan Wang
- The Department of Hematology and Oncology, The Second Hospital of Jilin University, China.
| | - Yan Sun
- The Department of Hematology and Oncology, The Second Hospital of Jilin University, China.
| | - Guang Shi
- The Department of Hematology and Oncology, The Second Hospital of Jilin University, China.
| | - Qiong Yu
- The Department of Hematology and Oncology, The Second Hospital of Jilin University, China.
| |
Collapse
|
34
|
Ruvolo PP. Galectins as regulators of cell survival in the leukemia niche. Adv Biol Regul 2018; 71:41-54. [PMID: 30245264 DOI: 10.1016/j.jbior.2018.09.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 09/10/2018] [Accepted: 09/11/2018] [Indexed: 02/08/2023]
Abstract
The microenvironment within the bone marrow (BM) contains support cells that promote leukemia cell survival and suppress host anti-tumor defenses. Galectins are a family of beta-galactoside binding proteins that are critical components in the tumor microenvironment. Galectin 1 (LGALS1) and Galectin 3 (LGALS3) as regulators of RAS signaling intracellularly and as inhibitors of immune cells extracellularly are perhaps the best studied members for their role in leukemia biology. Interest in Galectin 9 (LGALS9) is growing as this galectin has been identified as an immune checkpoint molecule. LGALS9 also supports leukemia stem cells (LSCs) though a mechanism of action is not clear. LGALS1 and LGALS3 each participate in a diverse number of survival pathways that promote drug resistance by supporting pro-tumor molecules such BCL2, MCL-1, and MYC and blocking tumor suppressors like p53. Acute myeloid leukemia (AML) BM mesenchymal stromal cells (MSC) have protein signatures that differ from healthy donor MSC. Elevated LGALS3 protein in AML MSC is associated with refractory disease/relapse demonstrating that MSC derived galectin impacts patient survival. LGALS3 is a critical determining factor whether MSC differentiate into adipocytes or osteoblasts so the galectin influences the cellular composition of the leukemia niche. Both LGALS3 and LGALS1 when secreted can suppress immune function. Both galectins can induce apoptosis of T cells. LGALS3 also modulates T cell receptor endocytosis and impairs interferon mediated chemokine production by binding glycosylated interferon. LGALS3 as a TIM3 binding partner acts to suppress T cell function. Galectins also impact leukemia cell mobilization and may participate in homing mechanisms. LGALS3 participates in transport mechanism of integrins, receptors, and other molecules that control cell adhesion and cell:cell interactions. The diversity of these various functions demonstrate the importance of these galectins in the leukemia niche. This review will cover the role of LGALS1, LGALS3, and LGALS9 in the various processes that are critical for maintaining leukemia cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Peter P Ruvolo
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
35
|
Moghaddam Y, Andalib A, Mohammad-Ganji M, Homayouni V, Sharifi M, Ganjalikhani-Hakemi M. Evaluation of the effect of TIM-3 suppression by miR-498 and its effect on apoptosis and proliferation rate of HL-60 cell line. Pathol Res Pract 2018; 214:1482-1488. [PMID: 30107988 DOI: 10.1016/j.prp.2018.07.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/29/2018] [Accepted: 07/22/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Acute Myeloid Leukemia (AML) is a Cancer of hematopoietic stem cells with a rapid progression. TIM-3 is expressed on leukemic stem cells (LSCs) in most types of AML and might have a positive effect on maintenance of malignant phenotype. MicroRNAs play important roles in either cancer progression or suppression. In this study were evaluated, the inhibitory effect of miR-498 on TIM-3 expression and its impact on proliferation and survival of HL-60 cell line. METHODS Firstly, the probable inhibitory effect of miR-498 on TIM-3 expression was predicted. HL-60 cells were cultured and expression of TIM-3 was induced on the cells using phorbol miristate acetate. The cells were transfected with miR-498 and expression level of TIM-3 were measured using with q-RT-PCR and flow cytometry methods. In addition, the effect of suppression of TIM-3 expression in HL-60 cell line was analyzed with apoptosis and cell proliferation assays. RESULTS Bioinformatics analyses predicted that miR-498 has high ability to silence TIM-3 gene expression. Our experiments confirmed that miR-498 was able to strongly silence TIM-3 expression (68% silencing) in HL-60 cell line (P < 0.002). Also, the cells with suppressed expression of TIM-3 had a lower proliferation and higher apoptosis rates. CONCLUSION Based on our results, the miR-498 can effectively suppress TIM-3 expression in the AML cell line. TIM-3 suppression, in turn, inhibits malignant cell proliferation and induces its apoptosis. Collectively, suppression of TIM-3 by miR-498 can be considered as a potential powerful way for treatment of AML.
Collapse
Affiliation(s)
- Yaser Moghaddam
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alireza Andalib
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Mohammad-Ganji
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Vida Homayouni
- Acqured Immunodeficiency Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammadreza Sharifi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Mazdak Ganjalikhani-Hakemi
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Acqured Immunodeficiency Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
36
|
Yasinska IM, Gonçalves Silva I, Sakhnevych S, Gibbs BF, Raap U, Fasler-Kan E, Sumbayev VV. Biochemical mechanisms implemented by human acute myeloid leukemia cells to suppress host immune surveillance. Cell Mol Immunol 2018; 15:989-991. [PMID: 29872115 DOI: 10.1038/s41423-018-0047-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 05/04/2018] [Indexed: 01/21/2023] Open
Abstract
Acute myeloid leukaemia (AML) is a blood/bone marrow cancer originating from myeloid cell precusors capable of self-renewing. AML cells implement biochemical mechanisms which allow them not only to survive, but also to successfully escape immune surveillance. ln this work, we discuss crucial molecular mechanisms used by human AML cells in order to evade immune attack.
Collapse
Affiliation(s)
- Inna M Yasinska
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, UK
| | | | - Svetlana Sakhnevych
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, UK
| | - Bernhard F Gibbs
- Department of Dermatology and Allergy, University of Oldenburg, Oldenburg, Germany
| | - Ulrike Raap
- Department of Dermatology and Allergy, University of Oldenburg, Oldenburg, Germany
| | - Elizaveta Fasler-Kan
- Department of Pediatric Surgery and Department of Biomedical Research, Children's Hospital, Inselspital, University of Bern, Bern, Switzerland.,Department of Biomedicine University Hospital Basel and University of Basel, Basel, Switzerland
| | - Vadim V Sumbayev
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, UK.
| |
Collapse
|
37
|
Yasinska IM, Gonçalves Silva I, Sakhnevych SS, Ruegg L, Hussain R, Siligardi G, Fiedler W, Wellbrock J, Bardelli M, Varani L, Raap U, Berger S, Gibbs BF, Fasler-Kan E, Sumbayev VV. High mobility group box 1 (HMGB1) acts as an "alarmin" to promote acute myeloid leukaemia progression. Oncoimmunology 2018; 7:e1438109. [PMID: 29872582 PMCID: PMC5980411 DOI: 10.1080/2162402x.2018.1438109] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 01/30/2018] [Accepted: 02/02/2018] [Indexed: 02/08/2023] Open
Abstract
High mobility group box 1 (HMGB1) is a non-histone protein localised in the cell nucleus, where it interacts with DNA and promotes nuclear transcription events. HMGB1 levels are elevated during acute myeloid leukaemia (AML) progression followed by participation of this protein in triggering signalling events in target cells as a pro-inflammatory stimulus. This mechanism was hypothesised to be employed as a survival pathway by malignant blood cells and our aims were therefore to test this hypothesis experimentally. Here we report that HMGB1 triggers the release of tumour necrosis factor alpha (TNF-α) by primary human AML cells. TNF-α induces interleukin 1 beta (IL-1β) production by healthy leukocytes, leading to IL-1β-induced secretion of stem cell factor (SCF) by competent cells (for example endothelial cells). These results were verified in mouse bone marrow and primary human AML blood plasma samples. In addition, HMGB1 was found to induce secretion of angiogenic vascular endothelial growth factor (VEGF) and this process was dependent on the immune receptor Tim-3. We therefore conclude that HMGB1 is critical for AML progression as a ligand of Tim-3 and other immune receptors thus supporting survival/proliferation of AML cells and possibly the process of angiogenesis.
Collapse
Affiliation(s)
- Inna M Yasinska
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - Isabel Gonçalves Silva
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - Svetlana S Sakhnevych
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - Laura Ruegg
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | | | | | - Walter Fiedler
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Germany
| | - Jasmin Wellbrock
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Germany
| | - Marco Bardelli
- Institute for Research in Biomedicine, Universita' della Svizzera italiana (USI), Bellinzona, Switzerland
| | - Luca Varani
- Institute for Research in Biomedicine, Universita' della Svizzera italiana (USI), Bellinzona, Switzerland
| | - Ulrike Raap
- Department of Medicine (Dermatology and Allergology), University of Oldenburg, Germany
| | - Steffen Berger
- Department of Pediatric Surgery and Department of Biomedical Research, Children's Hospital, Inselspital, University of Bern, Switzerland
| | - Bernhard F Gibbs
- Department of Medicine (Dermatology and Allergology), University of Oldenburg, Germany
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| | - Elizaveta Fasler-Kan
- Department of Pediatric Surgery and Department of Biomedical Research, Children's Hospital, Inselspital, University of Bern, Switzerland
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Vadim V Sumbayev
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, United Kingdom
| |
Collapse
|
38
|
Sumbayev VV, Gonçalves Silva I, Blackburn J, Gibbs BF, Yasinska IM, Garrett MD, Tonevitsky AG, Ushkaryov YA. Expression of functional neuronal receptor latrophilin 1 in human acute myeloid leukaemia cells. Oncotarget 2018; 7:45575-45583. [PMID: 27322212 PMCID: PMC5216743 DOI: 10.18632/oncotarget.10039] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/29/2016] [Indexed: 12/04/2022] Open
Abstract
Acute myeloid leukaemia (AML) is a blood cancer affecting cells of myeloid lineage. It is characterised by rapid growth of malignant leukocytes that accumulate in the bone marrow and suppress normal haematopoiesis. This systemic disease remains a serious medical burden worldwide. Characterisation of protein antigens specifically expressed by malignant cells, but not by healthy leukocytes, is vital for the diagnostics and targeted treatment of AML. Here we report, for the first time, that the neuronal receptor latrophilin-1 is expressed in human monocytic leukaemia cell lines and in primary human AML cells. However, it is absent in healthy leukocytes. Latrophilin-1 is functional in leukaemia cells tested, and its biosynthesis is controlled through the mammalian target of rapamycin (mTOR), a master regulator of myeloid cell translational pathways. Our findings demonstrate that latrophilin-1 could be considered as a novel biomarker of human AML, which offers potential new avenues for AML diagnosis and treatment.
Collapse
Affiliation(s)
- Vadim V Sumbayev
- School of Pharmacy, University of Kent, Chatham, Kent, ME4 4TB, United Kingdom
| | | | - Jennifer Blackburn
- School of Pharmacy, University of Kent, Chatham, Kent, ME4 4TB, United Kingdom
| | - Bernhard F Gibbs
- School of Pharmacy, University of Kent, Chatham, Kent, ME4 4TB, United Kingdom
| | - Inna M Yasinska
- School of Pharmacy, University of Kent, Chatham, Kent, ME4 4TB, United Kingdom
| | - Michelle D Garrett
- School of Biosciences, University of Kent, Canterbury, Kent, CT2 7NJ, United Kingdom
| | - Alexander G Tonevitsky
- Hertsen Moscow Oncology Research Institute, Branch of The National Medical Research Radiological Center, Ministry of Health of The Russian Federation, 125284, Moscow, Russian Federation
| | - Yuri A Ushkaryov
- School of Pharmacy, University of Kent, Chatham, Kent, ME4 4TB, United Kingdom
| |
Collapse
|
39
|
Xu L, Xu J, Ma S, Li X, Zhu M, Chen S, Han Y, Tang X, Fu Z, Qiu H, Yu J, Wu D, Wu X. High Tim-3 expression on AML blasts could enhance chemotherapy sensitivity. Oncotarget 2017; 8:102088-102096. [PMID: 29254227 PMCID: PMC5731937 DOI: 10.18632/oncotarget.22141] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/17/2017] [Indexed: 02/06/2023] Open
Abstract
T-cell immunoglobulin and mucin domain-containing molecule3 (Tim-3) represents a novel mechanism of T-cell dysfunction and exhaustion. Tim-3 has also been identified in various solid tumors. However, the role of Tim-3 expression on blast cells in acute myeloid leukemia (AML) is not well understood. In this study, we aimed to explore the role of Tim-3 in patients with de novo AML, and the correlation between Tim-3 and clinicopathological prognosis. The study cohort consisted of 76 patients with de novo non-M3 AML. These patients’ bone marrow samples were collected and then bone marrow mononuclear cells (BMCs) were isolated for flow cytometry to detect Tim-3 expression on blasts. According to FAB type, 76 diagnosed AML patients included in this study were: M0 (n=2), M1 (n=16), M2 (n=20), M4 (n=20), M5 (n=16), and M6 (n=2). A positive expression (>20%) of Tim-3 was found in 87% (66/76) of patients with AML. The average percentage of Tim-3(+) blasts in these AML patients was 58.26 ± 29.23%. Moreover, the frequency of Tim-3 high expression was higher in M4 patients than that in other AML patients according to FAB type (P=0.004). Tim-3 high expression was also closely associated with inv(16) (P=0.01) and C/EBPA mutation (P=0.03). The mutations of the following six genes, i.e., FLT3-ITD, NPM1, C-KIT, IDH1/IDH2, DNMT3A, were independent of the Tim-3 expression. Additionally, it is more likely to find higher levels of Tim-3 in the low-risk group than in the intermediate- and high-risk groups (P=0.02). The expression of Tim-3 was positively correlated with CD13 (r=0.36, P=0.001), CD34 (r=0.41, P=0.000), and CD7 (r=0.27, P=0.02) in AML patients. AML patients with high Tim-3 expression achieved significantly high complete remission (CR) rate (P=0.01), while their Tim-3 expression significantly decreased after CR (P=0.01). Blockade of Tim-3 expression on AML blasts significantly reduced the Idarubicin (IDA)-mediated suppression of cell growth and reduction of cell apoptosis in vitro. Collectively, our study suggests that high Tim-3 expression on AML blasts could enhances chemotherapy sensitivity.
Collapse
Affiliation(s)
- Liangjing Xu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Jinge Xu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China.,The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Shoubao Ma
- Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Xiaoli Li
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Mingqing Zhu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Suning Chen
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Yue Han
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Xiaowen Tang
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Zhengzheng Fu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Huiying Qiu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Jianhua Yu
- The Ohio State University, Comprehensive Cancer Center, Columbus, OH, USA
| | - Depei Wu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Xiaojin Wu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| |
Collapse
|
40
|
Marcucci F, Rumio C, Corti A. Tumor cell-associated immune checkpoint molecules - Drivers of malignancy and stemness. Biochim Biophys Acta Rev Cancer 2017; 1868:571-583. [PMID: 29056539 DOI: 10.1016/j.bbcan.2017.10.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 10/17/2017] [Accepted: 10/18/2017] [Indexed: 02/06/2023]
Abstract
Inhibitory or stimulatory immune checkpoint molecules are expressed on a sizeable fraction of tumor cells in different tumor types. It was thought that the main function of tumor cell-associated immune checkpoint molecules would be the modulation (down- or upregulation) of antitumor immune responses. In recent years, however, it has become clear that the expression of immune checkpoint molecules on tumor cells has important consequences on the biology of the tumor cells themselves. In particular, a causal relationship between the expression of these molecules and the acquisition of malignant traits has been demonstrated. Thus, immune checkpoint molecules have been shown to promote the epithelial-mesenchymal transition of tumor cells, the acquisition of tumor-initiating potential and resistance to apoptosis and antitumor drugs, as well as the propensity to disseminate and metastasize. Herein, we review this evidence, with a main focus on PD-L1, the most intensively investigated tumor cell-associated immune checkpoint molecule and for which most information is available. Then, we discuss more concisely other tumor cell-associated immune checkpoint molecules that have also been shown to induce the acquisition of malignant traits, such as PD-1, B7-H3, B7-H4, Tim-3, CD70, CD28, CD137, CD40 and CD47. Open questions in this field as well as some therapeutic approaches that can be derived from this knowledge, are also addressed.
Collapse
Affiliation(s)
- Fabrizio Marcucci
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Trentacoste 2, Milan, Italy.
| | - Cristiano Rumio
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Trentacoste 2, Milan, Italy.
| | - Angelo Corti
- Vita-Salute San Raffaele University, DIBIT-Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, via Olgettina 58, Milan, Italy.
| |
Collapse
|
41
|
Gonçalves Silva I, Yasinska IM, Sakhnevych SS, Fiedler W, Wellbrock J, Bardelli M, Varani L, Hussain R, Siligardi G, Ceccone G, Berger SM, Ushkaryov YA, Gibbs BF, Fasler-Kan E, Sumbayev VV. The Tim-3-galectin-9 Secretory Pathway is Involved in the Immune Escape of Human Acute Myeloid Leukemia Cells. EBioMedicine 2017; 22:44-57. [PMID: 28750861 PMCID: PMC5552242 DOI: 10.1016/j.ebiom.2017.07.018] [Citation(s) in RCA: 177] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/12/2017] [Accepted: 07/17/2017] [Indexed: 01/02/2023] Open
Abstract
Acute myeloid leukemia (AML) is a severe and often fatal systemic malignancy. Malignant cells are capable of escaping host immune surveillance by inactivating cytotoxic lymphoid cells. In this work we discovered a fundamental molecular pathway, which includes ligand-dependent activation of ectopically expressed latrophilin 1 and possibly other G-protein coupled receptors leading to increased translation and exocytosis of the immune receptor Tim-3 and its ligand galectin-9. This occurs in a protein kinase C and mTOR (mammalian target of rapamycin)-dependent manner. Tim-3 participates in galectin-9 secretion and is also released in a free soluble form. Galectin-9 impairs the anti-cancer activity of cytotoxic lymphoid cells including natural killer (NK) cells. Soluble Tim-3 prevents secretion of interleukin-2 (IL-2) required for the activation of cytotoxic lymphoid cells. These results were validated in ex vivo experiments using primary samples from AML patients. This pathway provides reliable targets for both highly specific diagnosis and immune therapy of AML.
Collapse
Affiliation(s)
| | - Inna M Yasinska
- School of Pharmacy, University of Kent, Chatham Maritime, UK
| | | | - Walter Fiedler
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Germany
| | - Jasmin Wellbrock
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Germany
| | - Marco Bardelli
- Institute for Research in Biomedicine, Universita' della Svizzera italiana (USI), Bellinzona, Switzerland
| | - Luca Varani
- Institute for Research in Biomedicine, Universita' della Svizzera italiana (USI), Bellinzona, Switzerland
| | | | | | | | - Steffen M Berger
- Department of Pediatric Surgery and Department of Clinical Research, Children's Hospital, Inselspital, University of Bern, Switzerland
| | | | - Bernhard F Gibbs
- School of Pharmacy, University of Kent, Chatham Maritime, UK; Department of Dermatology, University of Oldenburg, Germany.
| | - Elizaveta Fasler-Kan
- Department of Pediatric Surgery and Department of Clinical Research, Children's Hospital, Inselspital, University of Bern, Switzerland; Department of Biomedicine, University of Basel, Switzerland.
| | | |
Collapse
|
42
|
Das M, Zhu C, Kuchroo VK. Tim-3 and its role in regulating anti-tumor immunity. Immunol Rev 2017; 276:97-111. [PMID: 28258697 DOI: 10.1111/imr.12520] [Citation(s) in RCA: 628] [Impact Index Per Article: 78.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 11/19/2016] [Indexed: 12/13/2022]
Abstract
Immunotherapy is being increasingly recognized as a key therapeutic modality to treat cancer and represents one of the most exciting treatments for the disease. Fighting cancer with immunotherapy has revolutionized treatment for some patients and therapies targeting the immune checkpoint molecules such as CTLA-4 and PD-1 have achieved durable responses in melanoma, renal cancer, Hodgkin's diseases and lung cancer. However, the success rate of these treatments has been low and a large number of cancers, including colorectal cancer remain largely refractory to CTLA-4 and PD-1 blockade. This has provided impetus to identify other co-inhibitory receptors that could be exploited to enhance response rates of current immunotherapeutic agents and achieve responses to the cancers that are refectory to immunotherapy. Tim-3 is a co-inhibitory receptor that is expressed on IFN-g-producing T cells, FoxP3+ Treg cells and innate immune cells (macrophages and dendritic cells) where it has been shown to suppress their responses upon interaction with their ligand(s). Tim-3 has gained prominence as a potential candidate for cancer immunotherapy, where it has been shown that in vivo blockade of Tim-3 with other check-point inhibitors enhances anti-tumor immunity and suppresses tumor growth in several preclinical tumor models. This review discusses the recent findings on Tim-3, the role it plays in regulating immune responses in different cell types and the rationale for targeting Tim-3 for effective cancer immunotherapy.
Collapse
Affiliation(s)
- Madhumita Das
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA.,Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Chen Zhu
- Discovery Biology, Research and Development, Sanofi US, Cambridge, MA, USA
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA.,Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA.,The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
43
|
Gonçalves Silva I, Gibbs BF, Bardelli M, Varani L, Sumbayev VV. Differential expression and biochemical activity of the immune receptor Tim-3 in healthy and malignant human myeloid cells. Oncotarget 2016; 6:33823-33. [PMID: 26413815 PMCID: PMC4741805 DOI: 10.18632/oncotarget.5257] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 09/04/2015] [Indexed: 01/21/2023] Open
Abstract
The T cell immunoglobulin and mucin domain 3 (Tim-3) is a plasma membrane-associated receptor which is involved in a variety of biological responses in human immune cells. It is highly expressed in most acute myeloid leukaemia (AML) cells and therefore may serve as a possible target for AML therapy. However, its biochemical activities in primary human AML cells remain unclear. We therefore analysed the total expression and surface presence of the Tim-3 receptor in primary human AML blasts and healthy primary human leukocytes isolated from human blood. We found that Tim-3 expression was significantly higher in primary AML cells compared to primary healthy leukocytes. Tim-3 receptor molecules were distributed largely on the surface of primary AML cells, whereas in healthy leukocytes Tim-3 protein was mainly expressed intracellularly. In primary human AML blasts, both Tim-3 agonistic antibody and galectin-9 (a Tim-3 natural ligand) significantly upregulated mTOR pathway activity. This was in line with increased accumulation of hypoxia-inducible factor 1 alpha (HIF-1α) and secretion of VEGF and TNF-α. Similar results were obtained in primary human healthy leukocytes. Importantly, in both types of primary cells, Tim-3-mediated effects were compared with those induced by lipopolysaccharide (LPS) and stem cell factor (SCF). Tim-3 induced comparatively moderate responses in both AML cells and healthy leukocytes. However, Tim-3, like LPS, mediated the release of both TNF-α and VEGF, while SCF induced mostly VEGF secretion and did not upregulate TNF-α release.
Collapse
Affiliation(s)
| | - Bernhard F Gibbs
- School of Pharmacy, University of Kent, Kent, ME4 4TB, United Kingdom
| | - Marco Bardelli
- Institute for Research in Biomedicine, Universita' della Svizzera Italiana (USI) 6500 Bellinzona, Switzerland
| | - Luca Varani
- Institute for Research in Biomedicine, Universita' della Svizzera Italiana (USI) 6500 Bellinzona, Switzerland
| | - Vadim V Sumbayev
- School of Pharmacy, University of Kent, Kent, ME4 4TB, United Kingdom
| |
Collapse
|
44
|
Gibbs BF, Gonçalves Silva I, Prokhorov A, Abooali M, Yasinska IM, Casely-Hayford MA, Berger SM, Fasler-Kan E, Sumbayev VV. Caffeine affects the biological responses of human hematopoietic cells of myeloid lineage via downregulation of the mTOR pathway and xanthine oxidase activity. Oncotarget 2016; 6:28678-92. [PMID: 26384306 PMCID: PMC4745685 DOI: 10.18632/oncotarget.5212] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 08/31/2015] [Indexed: 01/07/2023] Open
Abstract
Correction of human myeloid cell function is crucial for the prevention of inflammatory and allergic reactions as well as leukaemia progression. Caffeine, a naturally occurring food component, is known to display anti-inflammatory effects which have previously been ascribed largely to its inhibitory actions on phosphodiesterase. However, more recent studies suggest an additional role in affecting the activity of the mammalian target of rapamycin (mTOR), a master regulator of myeloid cell translational pathways, although detailed molecular events underlying its mode of action have not been elucidated. Here, we report the cellular uptake of caffeine, without metabolisation, by healthy and malignant hematopoietic myeloid cells including monocytes, basophils and primary acute myeloid leukaemia mononuclear blasts. Unmodified caffeine downregulated mTOR signalling, which affected glycolysis and the release of pro-inflammatory/pro-angiogenic cytokines as well as other inflammatory mediators. In monocytes, the effects of caffeine were potentiated by its ability to inhibit xanthine oxidase, an enzyme which plays a central role in human purine catabolism by generating uric acid. In basophils, caffeine also increased intracellular cyclic adenosine monophosphate (cAMP) levels which further enhanced its inhibitory action on mTOR. These results demonstrate an important mode of pharmacological action of caffeine with potentially wide-ranging therapeutic impact for treating non-infectious disorders of the human immune system, where it could be applied directly to inflammatory cells.
Collapse
Affiliation(s)
- Bernhard F Gibbs
- School of Pharmacy, University of Kent, Chatham Maritime, ME4 4TB Kent, United Kingdom
| | | | - Alexandr Prokhorov
- School of Pharmacy, University of Kent, Chatham Maritime, ME4 4TB Kent, United Kingdom
| | - Maryam Abooali
- School of Pharmacy, University of Kent, Chatham Maritime, ME4 4TB Kent, United Kingdom
| | - Inna M Yasinska
- School of Pharmacy, University of Kent, Chatham Maritime, ME4 4TB Kent, United Kingdom
| | | | - Steffen M Berger
- Department of Pediatric Surgery and Department of Clinical Research, Inselspital, University Hospital, University of Bern, CH-3010 Bern, Switzerland
| | - Elizaveta Fasler-Kan
- Department of Pediatric Surgery and Department of Clinical Research, Inselspital, University Hospital, University of Bern, CH-3010 Bern, Switzerland.,Department of Biomedicine, University of Basel and University Hospital Basel, CH-4031 Basel, Switzerland
| | - Vadim V Sumbayev
- School of Pharmacy, University of Kent, Chatham Maritime, ME4 4TB Kent, United Kingdom
| |
Collapse
|
45
|
Gonçalves Silva I, Rüegg L, Gibbs BF, Bardelli M, Fruehwirth A, Varani L, Berger SM, Fasler-Kan E, Sumbayev VV. The immune receptor Tim-3 acts as a trafficker in a Tim-3/galectin-9 autocrine loop in human myeloid leukemia cells. Oncoimmunology 2016; 5:e1195535. [PMID: 27622049 PMCID: PMC5006895 DOI: 10.1080/2162402x.2016.1195535] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 05/19/2016] [Accepted: 05/19/2016] [Indexed: 01/31/2023] Open
Abstract
The immune receptor Tim-3 is often highly expressed in human acute myeloid leukemia (AML) cells where it acts as a growth factor and inflammatory receptor. Recently, it has been demonstrated that Tim-3 forms an autocrine loop with its natural ligand galectin-9 in human AML cells. However, the pathophysiological functions of Tim-3 in human AML cells remain unclear. Here, we report for the first time that Tim-3 is required for galectin-9 secretion in human AML cells. However, this effect is cell-type specific and was found so far to be applicable only to myeloid (and not, for example, lymphoid) leukemia cells. We concluded that AML cells might use Tim-3 as a trafficker for the secretion of galectin-9 which can then be possibly used to impair the anticancer activities of cytotoxic T cells and natural killer (NK) cells.
Collapse
Affiliation(s)
| | - Laura Rüegg
- School of Pharmacy, University of Kent , Canterbury, United Kingdom
| | - Bernhard F Gibbs
- School of Pharmacy, University of Kent , Canterbury, United Kingdom
| | - Marco Bardelli
- Institute for Research in Biomedicine, Universita' della Svizzera italiana (USI) , Bellinzona, Switzerland
| | - Alexander Fruehwirth
- Institute for Research in Biomedicine, Universita' della Svizzera italiana (USI) , Bellinzona, Switzerland
| | - Luca Varani
- Institute for Research in Biomedicine, Universita' della Svizzera italiana (USI) , Bellinzona, Switzerland
| | - Steffen M Berger
- Department of Pediatric Surgery and Department of Clinical Research, Children's Hospital, Inselspital, University of Bern , Bern, Switzerland
| | - Elizaveta Fasler-Kan
- Department of Pediatric Surgery and Department of Clinical Research, Children's Hospital, Inselspital, University of Bern, Bern, Switzerland; Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Vadim V Sumbayev
- School of Pharmacy, University of Kent , Canterbury, United Kingdom
| |
Collapse
|
46
|
Leukemia Stem Cell-Released Microvesicles Promote the Survival and Migration of Myeloid Leukemia Cells and These Effects Can Be Inhibited by MicroRNA34a Overexpression. Stem Cells Int 2016; 2016:9313425. [PMID: 27127521 PMCID: PMC4835649 DOI: 10.1155/2016/9313425] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/07/2016] [Indexed: 11/18/2022] Open
Abstract
Leukemia stem cells (LSCs) play the major role in relapse of acute myeloid leukemia (AML). Recent evidence indicates that microvesicles (MVs) released from cancer stem cells can promote tumor growth and invasion. In this study, we investigated whether LSCs-released MVs (LMVs) can regulate the malignance of AML cells and whether overexpression of tumor suppressive microRNA (miR), miR34a, is able to interrupt this process. LSCs were transfected with miRNA control (miRCtrl) or miR34a mimic for producing LMVs, respectively, defined as LMVsmiRCtrl and LMVsmiR34a. The effect of miR34a transfection on LSC proliferation and the effects of LMVsmiRCtrl or LMVsmiR34a on the proliferation, migration, and apoptosis of AML cells (after LSC depletion) were determined. The levels of miR34a targets, caspase-3 and T cell immunoglobulin mucin-3 (Tim-3), were analyzed. Results showed that (1) LMVsmiRCtrl promoted proliferation and migration and inhibited apoptosis of AML cells, which were associated with miR34a deficit; (2) transfection of miR34a mimic inhibited LSC proliferation and increased miR34a level in LMVsmiR34a; (3) LMVsmiR34a produced opposite effects as compared with LMVsmiRCtrl, which were associated with the changes of caspase-3 and Tim-3 levels. In summary, LMVs support AML cell malignance and modulating miR34a could offer a new approach for the management of AML.
Collapse
|
47
|
Pelosi E, Castelli G, Testa U. Targeting LSCs through membrane antigens selectively or preferentially expressed on these cells. Blood Cells Mol Dis 2015; 55:336-46. [PMID: 26460257 DOI: 10.1016/j.bcmd.2015.07.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 07/17/2015] [Indexed: 02/08/2023]
Abstract
Studies of xenotransplantation of bone marrow and blood cells of AML patients have supported the existence of rare leukemic stem cells, able to initiate and maintain the leukemic process and bearing the typical leukemic abnormalities. LSCs possess self-renewal capacity and are responsible for the growth of the more differentiated leukemic progeny in the bone marrow and in the blood. These cells are more resistant than bulk leukemic cells to anti-leukemic drugs, thus survive to treatment and are, at a large extent, responsible for leukemia relapse. During the last two decades, considerable progresses have been made in the understanding of the peculiar cellular and molecular properties of LSCs. In this context, particularly relevant was the discovery of several membrane markers, selectively or preferentially expressed on LSCs. These membrane markers offer now unique opportunities to identify LSCs and to distinguish them from normal HSCs, to monitor the response of the various anti-leukemic treatments at the level of the LSC compartment, to identify relevant therapeutic targets. Concerning this last point, the most promising therapeutic targets are CD33 and CD123.
Collapse
Affiliation(s)
- Elvira Pelosi
- Department of Hematology, Oncology and Molecular Medicine, Istituto Suepriore di Sanità, Rome, Italy
| | - Germana Castelli
- Department of Hematology, Oncology and Molecular Medicine, Istituto Suepriore di Sanità, Rome, Italy
| | - Ugo Testa
- Department of Hematology, Oncology and Molecular Medicine, Istituto Suepriore di Sanità, Rome, Italy
| |
Collapse
|
48
|
Nagahara K, Arikawa T, Oomizu S, Kontani K, Nobumoto A, Tateno H, Watanabe K, Niki T, Katoh S, Miyake M, Nagahata SI, Hirabayashi J, Kuchroo VK, Yamauchi A, Hirashima M. Galectin-9 increases Tim-3+ dendritic cells and CD8+ T cells and enhances antitumor immunity via galectin-9-Tim-3 interactions. THE JOURNAL OF IMMUNOLOGY 2008; 181:7660-9. [PMID: 19017954 DOI: 10.4049/jimmunol.181.11.7660] [Citation(s) in RCA: 171] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A Tim-3 ligand, galectin-9 (Gal-9), modulates various functions of innate and adaptive immune responses. In this study, we demonstrate that Gal-9 prolongs the survival of Meth-A tumor-bearing mice in a dose- and time-dependent manner. Although Gal-9 did not prolong the survival of tumor-bearing nude mice, transfer of naive spleen cells restored a prolonged Gal-9-induced survival in nude mice, indicating possible involvement of T cell-mediated immune responses in Gal-9-mediated antitumor activity. Gal-9 administration increased the number of IFN-gamma-producing Tim-3(+) CD8(+) T cells with enhanced granzyme B and perforin expression, although it induced CD4(+) T cell apoptosis. It simultaneously increased the number of Tim-3(+)CD86(+) mature dendritic cells (DCs) in vivo and in vitro. Coculture of CD8(+) T cells with DCs from Gal-9-treated mice increased the number of IFN-gamma producing cells and IFN-gamma production. Depletion of Tim-3(+) DCs from DCs of Gal-9-treated tumor-bearing mice decreased the number of IFN-gamma-producing CD8(+) T cells. Such DC activity was significantly abrogated by Tim-3-Ig, suggesting that Gal-9 potentiates CD8(+) T cell-mediated antitumor immunity via Gal-9-Tim-3 interactions between DCs and CD8(+) T cells.
Collapse
Affiliation(s)
- Keiko Nagahara
- Department of Immunology and Immunopathology, Kagawa University, Kita-gun, Kagawa, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|