1
|
Abdel-Rahman S, Ovchinnikov V, Gabr MT. Structure-Based Rational Design of Constrained Peptides as TIM-3 Inhibitors. ACS Med Chem Lett 2024; 15:806-813. [PMID: 38894912 PMCID: PMC11181482 DOI: 10.1021/acsmedchemlett.3c00567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 05/02/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Blocking the immunosuppressive function of T-cell immunoglobulin mucin-3 (TIM-3) is an established therapeutic strategy to maximize the efficacy of immune checkpoint inhibitors for cancer immunotherapy. Currently, effective inhibition of TIM-3 interactions relies on monoclonal antibodies (mAbs), which come with drawbacks such as immunogenicity risk, limited tumor penetration, and high manufacturing costs. Guided by the X-ray cocrystal structures of TIM-3 with mAbs, we report an in silico structure-based rational design of constrained peptides as potent TIM-3 inhibitors. The top cyclic peptide from our study (P2) binds TIM-3 with a K D value of 166.3 ± 12.1 nM as determined by surface plasmon resonance (SPR) screening. Remarkably, P2 efficiently inhibits key TIM-3 interactions with natural TIM-3 ligands at submicromolar concentrations in a panel of cell-free and cell-based assays. The capacity of P2 to reverse immunosuppression in T-cell/cancer cell cocultures, coupled with favorable in vitro pharmacokinetic properties, highlights the potential of P2 for further evaluation in preclinical models of immuno-oncology.
Collapse
Affiliation(s)
- Somaya
A. Abdel-Rahman
- Department
of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, New York 10065, United States
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Victor Ovchinnikov
- Department
of Chemistry and Chemical Biology, Harvard
University, Cambridge, Massachusetts 02138, United States
| | - Moustafa T. Gabr
- Department
of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, New York 10065, United States
| |
Collapse
|
2
|
Tutunea-Fatan E, Arumugarajah S, Suri RS, Edgar CR, Hon I, Dikeakos JD, Gunaratnam L. Sensing Dying Cells in Health and Disease: The Importance of Kidney Injury Molecule-1. J Am Soc Nephrol 2024; 35:795-808. [PMID: 38353655 PMCID: PMC11164124 DOI: 10.1681/asn.0000000000000334] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024] Open
Abstract
Kidney injury molecule-1 (KIM-1), also known as T-cell Ig and mucin domain-1 (TIM-1), is a widely recognized biomarker for AKI, but its biological function is less appreciated. KIM-1/TIM-1 belongs to the T-cell Ig and mucin domain family of conserved transmembrane proteins, which bear the characteristic six-cysteine Ig-like variable domain. The latter enables binding of KIM-1/TIM-1 to its natural ligand, phosphatidylserine, expressed on the surface of apoptotic cells and necrotic cells. KIM-1/TIM-1 is expressed in a variety of tissues and plays fundamental roles in regulating sterile inflammation and adaptive immune responses. In the kidney, KIM-1 is upregulated on injured renal proximal tubule cells, which transforms them into phagocytes for clearance of dying cells and helps to dampen sterile inflammation. TIM-1, expressed in T cells, B cells, and natural killer T cells, is essential for cell activation and immune regulatory functions in the host. Functional polymorphisms in the gene for KIM-1/TIM-1, HAVCR1 , have been associated with susceptibility to immunoinflammatory conditions and hepatitis A virus-induced liver failure, which is thought to be due to a differential ability of KIM-1/TIM-1 variants to bind phosphatidylserine. This review will summarize the role of KIM-1/TIM-1 in health and disease and its potential clinical applications as a biomarker and therapeutic target in humans.
Collapse
Affiliation(s)
- Elena Tutunea-Fatan
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada
| | - Shabitha Arumugarajah
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Rita S. Suri
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Division of Nephrology, Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Cassandra R. Edgar
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Ingrid Hon
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Jimmy D. Dikeakos
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Lakshman Gunaratnam
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Division of Nephrology, Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
3
|
Brozat JF, Harbalioğlu N, Hohlstein P, Abu Jhaisha S, Pollmanns MR, Adams JK, Wirtz TH, Hamesch K, Yagmur E, Weiskirchen R, Tacke F, Trautwein C, Koch A. Elevated Serum KIM-1 in Sepsis Correlates with Kidney Dysfunction and the Severity of Multi-Organ Critical Illness. Int J Mol Sci 2024; 25:5819. [PMID: 38892009 PMCID: PMC11172102 DOI: 10.3390/ijms25115819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/24/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
The kidney injury molecule (KIM)-1 is shed from proximal tubular cells in acute kidney injury (AKI), relaying tubular epithelial proliferation. Additionally, KIM-1 portends complex immunoregulation and is elevated after exposure to lipopolysaccharides. It thus may represent a biomarker in critical illness, sepsis, and sepsis-associated AKI (SA-AKI). To characterise and compare KIM-1 in these settings, we analysed KIM-1 serum concentrations in 192 critically ill patients admitted to the intensive care unit. Irrespective of kidney dysfunction, KIM-1 serum levels were significantly higher in patients with sepsis compared with other critical illnesses (191.6 vs. 132.2 pg/mL, p = 0.019) and were highest in patients with urogenital sepsis, followed by liver failure. Furthermore, KIM-1 levels were significantly elevated in critically ill patients who developed AKI within 48 h (273.3 vs. 125.8 pg/mL, p = 0.026) or later received renal replacement therapy (RRT) (299.7 vs. 146.3 pg/mL, p < 0.001). KIM-1 correlated with markers of renal function, inflammatory parameters, hematopoietic function, and cholangiocellular injury. Among subcomponents of the SOFA score, KIM-1 was elevated in patients with hyperbilirubinaemia (>2 mg/dL, p < 0.001) and thrombocytopenia (<150/nL, p = 0.018). In univariate and multivariate regression analyses, KIM-1 predicted sepsis, the need for RRT, and multi-organ dysfunction (MOD, SOFA > 12 and APACHE II ≥ 20) on the day of admission, adjusting for relevant comorbidities, bilirubin, and platelet count. Additionally, KIM-1 in multivariate regression was able to predict sepsis in patients without prior (CKD) or present (AKI) kidney injury. Our study suggests that next to its established role as a biomarker in kidney dysfunction, KIM-1 is associated with sepsis, biliary injury, and critical illness severity. It thus may offer aid for risk stratification in these patients.
Collapse
Affiliation(s)
- Jonathan Frederik Brozat
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
- Department of Hepatology and Gastroenterology, Charité—Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Augustenburger Platz 1, 13353 Berlin, Germany;
| | - Neval Harbalioğlu
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Philipp Hohlstein
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Samira Abu Jhaisha
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Maike Rebecca Pollmanns
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Jule Katharina Adams
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Theresa Hildegard Wirtz
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Karim Hamesch
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Eray Yagmur
- Institute of Laboratory Medicine, Western Palatine Hospital, Hellmut-Hartert-Straße 1, 67655 Kaiserslautern, Germany;
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, University Hospital Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany;
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité—Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Augustenburger Platz 1, 13353 Berlin, Germany;
| | - Christian Trautwein
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Alexander Koch
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| |
Collapse
|
4
|
Delova A, Pasc A, Monari A. Interaction of the Immune System TIM-3 Protein with a Model Cellular Membrane Containing Phosphatidyl-Serine Lipids. Chemistry 2024; 30:e202304318. [PMID: 38345892 DOI: 10.1002/chem.202304318] [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] [Indexed: 02/29/2024]
Abstract
T cell transmembrane, Immunoglobulin, and Mucin (TIM) are important immune system proteins which are especially present in T-cells and regulated the immune system by sensing cell engulfment and apoptotic processes. Their role is exerted by the capacity to detect the presence of phosphatidyl-serine lipid polar head in the outer leaflet of cellular membranes (correlated with apoptosis). In this contribution by using equilibrium and enhanced sampling molecular dynamics simulation we unravel the molecular bases and the thermodynamics of TIM, and in particular TIM-3, interaction with phosphatidyl serine in a lipid bilayer. Since TIM-3 deregulation is an important factor of pro-oncogenic tumor micro-environment understanding its functioning at a molecular level may pave the way to the development of original immunotherapeutic approaches.
Collapse
Affiliation(s)
| | - Andreea Pasc
- Université de Lorraine and CNRS, UMR 7053L2CM, F-54000, Nancy, France
| | - Antonio Monari
- Université Paris Cité and CNRS, ITDODYS, F-75006, Paris, France
| |
Collapse
|
5
|
Crimini E, Boscolo Bielo L, Berton Giachetti PPM, Pellizzari G, Antonarelli G, Taurelli Salimbeni B, Repetto M, Belli C, Curigliano G. Beyond PD(L)-1 Blockade in Microsatellite-Instable Cancers: Current Landscape of Immune Co-Inhibitory Receptor Targeting. Cancers (Basel) 2024; 16:281. [PMID: 38254772 PMCID: PMC10813411 DOI: 10.3390/cancers16020281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
High microsatellite instability (MSI-H) derives from genomic hypermutability due to deficient mismatch repair function. Colorectal (CRC) and endometrial cancers (EC) are the tumor types that more often present MSI-H. Anti-PD(L)-1 antibodies have been demonstrated to be agnostically effective in patients with MSI-H cancer, but 50-60% of them do not respond to single-agent treatment, highlighting the necessity of expanding their treatment opportunities. Ipilimumab (anti-CTLA4) is the only immune checkpoint inhibitor (ICI) non-targeting PD(L)-1 that has been approved so far by the FDA for MSI-H cancer, namely, CRC in combination with nivolumab. Anti-TIM3 antibody LY3321367 showed interesting clinical activity in combination with anti-PDL-1 antibody in patients with MSI-H cancer not previously treated with anti-PD(L)-1. In contrast, no clinical evidence is available for anti-LAG3, anti-TIGIT, anti-BTLA, anti-ICOS and anti-IDO1 antibodies in MSI-H cancers, but clinical trials are ongoing. Other immunotherapeutic strategies under study for MSI-H cancers include vaccines, systemic immunomodulators, STING agonists, PKM2 activators, T-cell immunotherapy, LAIR-1 immunosuppression reversal, IL5 superagonists, oncolytic viruses and IL12 partial agonists. In conclusion, several combination therapies of ICIs and novel strategies are emerging and may revolutionize the treatment paradigm of MSI-H patients in the future. A huge effort will be necessary to find reliable immune biomarkers to personalize therapeutical decisions.
Collapse
Affiliation(s)
- Edoardo Crimini
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy (G.P.); (G.A.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Luca Boscolo Bielo
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy (G.P.); (G.A.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Pier Paolo Maria Berton Giachetti
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy (G.P.); (G.A.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Gloria Pellizzari
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy (G.P.); (G.A.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Gabriele Antonarelli
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy (G.P.); (G.A.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Beatrice Taurelli Salimbeni
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy (G.P.); (G.A.)
| | - Matteo Repetto
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Carmen Belli
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy (G.P.); (G.A.)
| | - Giuseppe Curigliano
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy (G.P.); (G.A.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| |
Collapse
|
6
|
Abdel-Rahman SA, Talagayev V, Pach S, Wolber G, Gabr MT. Discovery of Small-Molecule TIM-3 Inhibitors for Acute Myeloid Leukemia Using Pharmacophore-Based Virtual Screening. J Med Chem 2023; 66:11464-11475. [PMID: 37566998 DOI: 10.1021/acs.jmedchem.3c00960] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2023]
Abstract
T-cell immunoglobulin and mucin domain 3 (TIM-3) is a negative immune checkpoint that represents a promising target for cancer immunotherapy. Although encouraging results have been observed for TIM-3 inhibition in the context of acute myeloid leukemia (AML), targeting TIM-3 is currently restricted to monoclonal antibodies (mAbs). To fill this gap, we implemented a pharmacophore-based screening approach to identify small-molecule TIM-3 inhibitors. Our approach resulted in the identification of hit compounds with TIM-3 binding affinity. Subsequently, we used the structure-activity relationship (SAR) by a catalog approach to identify compound A-41 with submicromolar TIM-3 binding affinity. Remarkably, A-41 demonstrated the ability to block TIM-3 interactions with key ligands and inhibited the immunosuppressive function of TIM-3 using an in vitro coculture assay. This work will pave the way for future drug discovery efforts aiming at the development of small-molecule inhibitors TIM-3 for AML.
Collapse
Affiliation(s)
- Somaya A Abdel-Rahman
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, New York 10065, United States
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Valerij Talagayev
- Institute of Pharmacy, Pharmaceutical and Medicinal Chemistry (Computer-Aided Drug Design), Freie Universitaet Berlin, Berlin 14195, Germany
| | - Szymon Pach
- Institute of Pharmacy, Pharmaceutical and Medicinal Chemistry (Computer-Aided Drug Design), Freie Universitaet Berlin, Berlin 14195, Germany
| | - Gerhard Wolber
- Institute of Pharmacy, Pharmaceutical and Medicinal Chemistry (Computer-Aided Drug Design), Freie Universitaet Berlin, Berlin 14195, Germany
| | - Moustafa T Gabr
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, New York 10065, United States
| |
Collapse
|
7
|
Zheng C, Shi Y, Zou Y. T cell co-stimulatory and co-inhibitory pathways in atopic dermatitis. Front Immunol 2023; 14:1081999. [PMID: 36993982 PMCID: PMC10040887 DOI: 10.3389/fimmu.2023.1081999] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/28/2023] [Indexed: 03/14/2023] Open
Abstract
The use of immune checkpoint inhibitors (ICIs) targeting the T cell inhibitory pathways has revolutionized cancer treatment. However, ICIs might induce progressive atopic dermatitis (AD) by affecting T cell reactivation. The critical role of T cells in AD pathogenesis is widely known. T cell co-signaling pathways regulate T cell activation, where co-signaling molecules are essential for determining the magnitude of the T cell response to antigens. Given the increasing use of ICIs in cancer treatment, a timely overview of the role of T cell co-signaling molecules in AD is required. In this review, we emphasize the importance of these molecules involved in AD pathogenesis. We also discuss the potential of targeting T cell co-signaling pathways to treat AD and present the unresolved issues and existing limitations. A better understanding of the T cell co-signaling pathways would aid investigation of the mechanism, prognosis evaluation, and treatment of AD.
Collapse
Affiliation(s)
- Chunjiao Zheng
- Skin and Cosmetic Research Department, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuling Shi
- Institute of Psoriasis, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Yuling Shi, ; Ying Zou,
| | - Ying Zou
- Skin and Cosmetic Research Department, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Yuling Shi, ; Ying Zou,
| |
Collapse
|
8
|
Mohd Idris RA, Mussa A, Ahmad S, Al-Hatamleh MAI, Hassan R, Tengku Din TADAA, Wan Abdul Rahman WF, Lazim NM, Boer JC, Plebanski M, Mohamud R. The Effects of Tamoxifen on Tolerogenic Cells in Cancer. BIOLOGY 2022; 11:1225. [PMID: 36009853 PMCID: PMC9405160 DOI: 10.3390/biology11081225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/01/2022] [Accepted: 08/01/2022] [Indexed: 11/30/2022]
Abstract
Tamoxifen (TAM) is the most prescribed selective estrogen receptor modulator (SERM) to treat hormone-receptor-positive breast cancer patients and has been used for more than 20 years. Its role as a hormone therapy is well established; however, the potential role in modulating tolerogenic cells needs to be better clarified. Infiltrating tumor-microenvironment-regulatory T cells (TME-Tregs) are important as they serve a suppressive function through the transcription factor Forkhead box P3 (Foxp3). Abundant studies have suggested that Foxp3 regulates the expression of several genes (CTLA-4, PD-1, LAG-3, TIM-3, TIGIT, TNFR2) involved in carcinogenesis to utilize its tumor suppressor function through knockout models. TAM is indirectly concomitant via the Cre/loxP system by allowing nuclear translocation of the fusion protein, excision of the floxed STOP cassette and heritable expression of encoding fluorescent protein in a cohort of cells that express Foxp3. Moreover, TAM administration in breast cancer treatment has shown its effects directly through MDSCs by the enrichment of its leukocyte populations, such as NK and NKT cells, while it impairs the differentiation and activation of DCs. However, the fundamental mechanisms of the reduction of this pool by TAM are unknown. Here, we review the vital effects of TAM on Tregs for a precise mechanistic understanding of cancer immunotherapies.
Collapse
Affiliation(s)
- Ros Akmal Mohd Idris
- Immunology Department, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Ali Mussa
- Haematology Department, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
- Department of Biology, Faculty of Education, Omdurman Islamic University, Omdurman P.O. Box 382, Sudan
| | - Suhana Ahmad
- Immunology Department, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Mohammad A. I. Al-Hatamleh
- Immunology Department, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Rosline Hassan
- Haematology Department, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | | | - Wan Faiziah Wan Abdul Rahman
- Pathology Department, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Norhafiza Mat Lazim
- Otorhinolaryngology Department-Head & Neck Surgery, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Jennifer C. Boer
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| | - Rohimah Mohamud
- Immunology Department, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| |
Collapse
|
9
|
Zhang M, Wang X, Hu L, Zhang Y, Zheng H, Wu H, Wang J, Luo L, Xiao H, Qiao C, Li X, Huang W, Wang Y, Feng J, Chen G. TIM-1 Augments Cellular Entry of Ebola Virus Species and Mutants, Which Is Blocked by Recombinant TIM-1 Protein. Microbiol Spectr 2022; 10:e0221221. [PMID: 35384693 PMCID: PMC9241846 DOI: 10.1128/spectrum.02212-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/07/2022] [Indexed: 01/12/2023] Open
Abstract
Ebola virus, a member of the Filoviridae family, utilizes the attachment factors on host cells to support its entry and cause severe tissue damage. TIM-1 has been identified as a predominant attachment factor via interaction with phosphatidylserine (PS) localized on the viral envelope and glycoprotein (GP). In this study, we give the first demonstration that TIM-1 enhances the cellular entry of three species of Ebola virus, as well as those harboring GP mutations (A82V, T544I, and A82V T544I). Furthermore, two TIM-1 variants (i.e., TIM-1-359aa and TIM-1-364aa) had comparable effects on promoting Zaire Ebola virus (EBOV) attachment, internalization, and infection. Importantly, recombinant TIM-1 ectodomain (ECD) protein could decrease the infectivity of Ebola virus and display synergistic inhibitory effects with ADI-15946, a monoclonal antibody with broad neutralizing activity to Ebola virus. Of note, EBOV strains harboring GP mutations (K510E and D552N), which were refractory to antibody treatment, were still sensitive to TIM-1 protein-mediated impairment of infectivity, indicating that TIM-1 protein may represent an alternative therapeutic regimen when antibody evasion occurs. IMPORTANCE The viral genome has acquired numerous mutations with the potential to increase transmission during the 2013-to-2016 outbreak of Ebola virus. EBOV strains harboring GP mutations (A82V, T544I, and A82V T544I), which have been identified to increase viral infectivity in humans, have attracted our attention. Herein, we give the first report that polymorphic TIM-1 enhances the infectivity of three species of Ebola virus, as well as those harboring GP mutations (A82V, T544I, and A82V T544I). We show that recombinant TIM-1 ECD protein could decrease the infectivity of Ebola virus with or without a point mutation and displays synergistic inhibitory effects with ADI-15946. Furthermore, TIM-1 protein potently blocked cell entry of antibody-evading Ebola virus species. These findings highlight the role of TIM-1 in Ebola virus infection and indicate that TIM-1 protein represents a potential therapeutic avenue for Ebola virus and its mutated species.
Collapse
Affiliation(s)
- Min Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Xinwei Wang
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, China
| | - Linhan Hu
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, China
| | - Yuting Zhang
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, China
| | - Hang Zheng
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, China
| | - Haiyan Wu
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Jing Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Longlong Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - He Xiao
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Chunxia Qiao
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Xinying Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Youchun Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Jiannan Feng
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Guojiang Chen
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
10
|
Xia M, Hu X, Zhao Q, Ru Y, Wang H, Zheng F. Development and Characterization of a Nanobody against Human T-Cell Immunoglobulin and Mucin-3. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:2929605. [PMID: 35726228 PMCID: PMC9206550 DOI: 10.1155/2022/2929605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/07/2022] [Accepted: 04/19/2022] [Indexed: 11/17/2022]
Abstract
Monoclonal antibodies and antibody-derived biologics are essential tools for cancer research and therapy. The development of monoclonal antibody treatments for successful tumor-targeted therapies took several decades. A nanobody constructed by molecular engineering of heavy-chain-only antibody, which is unique in camel or alpaca, is a burgeoning tools of diagnostic and therapeutic in clinic. In this study, we immunized a 4-year-old female alpaca with TIM-3 antigen. Then, a VHH phage was synthesized from the transcriptome of its B cells by nested PCR as an intermediate library; the library selection for Tim-3 antigen is carried out in three rounds of translation. The most reactive colonies were selected by periplasmic extract monoclonal ELISA. The nanobody was immobilized by metal affinity chromatography (IMAC) purification with the use of a Ni-NTA column, SDS-PAGE, and Western blotting. Finally, the affinity of TIM3-specific nanobody was determined by ELISA. As results, specific 15 kD bands representing nanomaterials were observed on the gel and confirmed by Western blotting. The nanobody showed obvious specific immune response to Tim-3 and had high binding affinity. We have successfully prepared a functional anti-human Tim-3 nanobody with high affinity in vitro.
Collapse
Affiliation(s)
- Mingyuan Xia
- Department of Urology, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an City, 710032 Shaanxi Province, China
| | - Xiangnan Hu
- No. 986 Hospital, Air Force Military Medical University, Xi'an City, 710054 Shaanxi Province, China
| | - Qiuxiang Zhao
- Department of Urology, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an City, 710032 Shaanxi Province, China
| | - Yi Ru
- Department of Biochemistry and Molecular Biology, Basic Medical College, Air Force Military Medical University, Xi'an City, 710032 Shaanxi Province, China
| | - He Wang
- Department of Urology, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an City, 710032 Shaanxi Province, China
| | - Fang Zheng
- The Key Laboratory of Environment and Genes Related to Disease of Ministry of Education, Health Science Center, Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
11
|
Carlino F, Diana A, Piccolo A, Ventriglia A, Bruno V, De Santo I, Letizia O, De Vita F, Daniele B, Ciardiello F, Orditura M. Immune-Based Therapy in Triple-Negative Breast Cancer: From Molecular Biology to Clinical Practice. Cancers (Basel) 2022; 14:cancers14092102. [PMID: 35565233 PMCID: PMC9103968 DOI: 10.3390/cancers14092102] [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: 02/17/2022] [Revised: 04/16/2022] [Accepted: 04/19/2022] [Indexed: 12/19/2022] Open
Abstract
Triple-negative breast cancer (TNBC) has been considered for many years an orphan disease in terms of therapeutic options, with conventional chemotherapy (CT) still representing the mainstay of treatment in the majority of patients. Although breast cancer (BC) has been historically considered a "cold tumor", exciting progress in the genomic field leading to the characterization of the molecular portrait and the immune profile of TNBC has opened the door to novel therapeutic strategies, including Immune Checkpoint Inhibitors (ICIs), Poly ADP-Ribose Polymerase (PARP) inhibitors and Antibody Drug Conjugates (ADCs). In particular, compared to standard CT, the immune-based approach has been demonstrated to improve progression-free survival (PFS) and overall survival (OS) in metastatic PD-L1-positive TNBC and the pathological complete response rate in the early setting, regardless of PD-L1 expression. To date, PD-L1 has been widely used as a predictor of the response to ICIs; however, many patients do not benefit from the addition of immunotherapy. Therefore, PD-L1 is not a reliable predictive biomarker of the response, and its accuracy remains controversial due to the lack of a consensus about the assay, the antibody, and the scoring system to adopt, as well as the spatial and temporal heterogeneity of the PD-L1 status. In the precision medicine era, there is an urgent need to identify more sensitive biomarkers in the BC immune oncology field other than just PD-L1 expression. Through the characterization of the tumor microenvironment (TME), the analysis of peripheral blood and the evaluation of immune gene signatures, novel potential biomarkers have been explored, such as the Tumor Mutational Burden (TMB), Microsatellite Instability/Mismatch Repair Deficiency (MSI/dMMR) status, genomic and epigenomic alterations and tumor-infiltrating lymphocytes (TILs). This review aims to summarize the recent knowledge on BC immunograms and on the biomarkers proposed to support ICI-based therapy in TNBC, as well as to provide an overview of the potential strategies to enhance the immune response in order to overcome the mechanisms of resistance.
Collapse
Affiliation(s)
- Francesca Carlino
- Department of Precision Medicine, Division of Medical Oncology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy; (A.P.); (A.V.); (V.B.); (F.D.V.); (F.C.); (M.O.)
- Medical Oncology Unit, Ospedale Ave Gratia Plena, San Felice a Cancello, 81027 Caserta, Italy; (I.D.S.); (O.L.)
- Correspondence: ; Tel.: +39-349-5152216
| | - Anna Diana
- Medical Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (A.D.); (B.D.)
| | - Antonio Piccolo
- Department of Precision Medicine, Division of Medical Oncology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy; (A.P.); (A.V.); (V.B.); (F.D.V.); (F.C.); (M.O.)
| | - Anna Ventriglia
- Department of Precision Medicine, Division of Medical Oncology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy; (A.P.); (A.V.); (V.B.); (F.D.V.); (F.C.); (M.O.)
| | - Vincenzo Bruno
- Department of Precision Medicine, Division of Medical Oncology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy; (A.P.); (A.V.); (V.B.); (F.D.V.); (F.C.); (M.O.)
| | - Irene De Santo
- Medical Oncology Unit, Ospedale Ave Gratia Plena, San Felice a Cancello, 81027 Caserta, Italy; (I.D.S.); (O.L.)
| | - Ortensio Letizia
- Medical Oncology Unit, Ospedale Ave Gratia Plena, San Felice a Cancello, 81027 Caserta, Italy; (I.D.S.); (O.L.)
| | - Ferdinando De Vita
- Department of Precision Medicine, Division of Medical Oncology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy; (A.P.); (A.V.); (V.B.); (F.D.V.); (F.C.); (M.O.)
| | - Bruno Daniele
- Medical Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (A.D.); (B.D.)
| | - Fortunato Ciardiello
- Department of Precision Medicine, Division of Medical Oncology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy; (A.P.); (A.V.); (V.B.); (F.D.V.); (F.C.); (M.O.)
| | - Michele Orditura
- Department of Precision Medicine, Division of Medical Oncology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy; (A.P.); (A.V.); (V.B.); (F.D.V.); (F.C.); (M.O.)
| |
Collapse
|
12
|
Rietz TA, Teuscher KB, Mills JJ, Gogliotti RD, Lepovitz LT, Scaggs WR, Yoshida K, Luong K, Lee T, Fesik SW. Fragment-Based Discovery of Small Molecules Bound to T-Cell Immunoglobulin and Mucin Domain-Containing Molecule 3 (TIM-3). J Med Chem 2021; 64:14757-14772. [PMID: 34597046 DOI: 10.1021/acs.jmedchem.1c01336] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
T-cell immunoglobulin and mucin domain-containing molecule 3 (TIM-3; HAVCR2) has emerged as an attractive immune checkpoint target for cancer immunotherapy. TIM-3 is a negative regulator of the systemic immune response to cancer and is expressed on several dysfunctional, or exhausted, immune cell subsets. Upregulation of TIM-3 is associated with tumor progression, poor survival rates, and acquired resistance to antibody-based immunotherapies in the clinic. Despite the potential advantages of small-molecule inhibitors over antibodies, the discovery of small-molecule inhibitors has lagged behind that of antibody therapeutics. Here, we describe the discovery of high-affinity small-molecule ligands for TIM-3 through an NMR-based fragment screen and structure-based lead optimization. These compounds represent useful tools to further study the biology of TIM-3 immune modulation in cancer and serve as a potentially useful starting point toward the discovery of TIM-3-targeted therapeutics.
Collapse
Affiliation(s)
- Tyson A Rietz
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States
| | - Kevin B Teuscher
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States
| | - Jonathan J Mills
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States
| | - Rocco D Gogliotti
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States
| | - Lance T Lepovitz
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States
| | - W Rush Scaggs
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States
| | - Keisuke Yoshida
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States
| | - Kelvin Luong
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-6600, United States
| | - Taekyu Lee
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States
| | - Stephen W Fesik
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-6600, United States.,Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| |
Collapse
|
13
|
Karmakova ТА, Sergeeva NS, Kanukoev КY, Alekseev BY, Kaprin АD. Kidney Injury Molecule 1 (KIM-1): a Multifunctional Glycoprotein and Biological Marker (Review). Sovrem Tekhnologii Med 2021; 13:64-78. [PMID: 34603757 PMCID: PMC8482821 DOI: 10.17691/stm2021.13.3.08] [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: 08/21/2020] [Indexed: 12/17/2022] Open
Abstract
KIM-1 (kidney injury molecule 1) is a transmembrane glycoprotein also known as HAVcr-1 and TIM-1 belongs to the T-cell immunoglobulin and mucin domain family (TIM) of proteins. TIM glycoproteins are presented on the immune cells and participate in the regulation of immune reactions. KIM-1 differs from other members of its family in that it is expressed not only by immunocompetent cells but epithelial cells as well. Cellular and humoral effects mediated by KIM-1 are involved in a variety of physiological and pathophysiological processes. Current understanding of the mechanisms determining the participation of KIM-1 in viral invasion, the immune response regulation, adaptive reactions of the kidney epithelium to acute ischemic or toxic injury, in progression of chronic renal diseases, and kidney cancer development have been presented in this review. Data of clinical researches demonstrating the association of KIM-1 with viral diseases and immune disorders have also been analyzed. Potential application of KIM-1 as urinary or serological marker in renal and cardiovascular diseases has been considered.
Collapse
Affiliation(s)
- Т А Karmakova
- Leading Researcher, Department of Predicting the Effectiveness of Conservative Therapy; P. Hertsen Moscow Oncology Research Institute - Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 3, 2 Botkinsky Proezd, Moscow, 125284, Russia
| | - N S Sergeeva
- Professor, Head of the Department of Predicting the Effectiveness of Conservative Therapy; P. Hertsen Moscow Oncology Research Institute - Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 3, 2 Botkinsky Proezd, Moscow, 125284, Russia; Professor, Department of Biology; Pirogov Russian National Research Medical University, 1 Ostrovitianova St., Moscow, 117997, Russia
| | - К Yu Kanukoev
- Urologist, Department of Urology with Chemotherapy; P. Hertsen Moscow Oncology Research Institute - Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 3, 2 Botkinsky Proezd, Moscow, 125284, Russia
| | - B Ya Alekseev
- Professor, Deputy General Director for Science; National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 4 Koroleva St., Obninsk, 249036, Russia
| | - А D Kaprin
- Professor, Academician of the Russian Academy of Sciences, General Director; National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 4 Koroleva St., Obninsk, 249036, Russia
| |
Collapse
|
14
|
Kirui J, Abidine Y, Lenman A, Islam K, Gwon YD, Lasswitz L, Evander M, Bally M, Gerold G. The Phosphatidylserine Receptor TIM-1 Enhances Authentic Chikungunya Virus Cell Entry. Cells 2021; 10:cells10071828. [PMID: 34359995 PMCID: PMC8303237 DOI: 10.3390/cells10071828] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 11/26/2022] Open
Abstract
Chikungunya virus (CHIKV) is a re-emerging, mosquito-transmitted, enveloped positive stranded RNA virus. Chikungunya fever is characterized by acute and chronic debilitating arthritis. Although multiple host factors have been shown to enhance CHIKV infection, the molecular mechanisms of cell entry and entry factors remain poorly understood. The phosphatidylserine-dependent receptors, T-cell immunoglobulin and mucin domain 1 (TIM-1) and Axl receptor tyrosine kinase (Axl), are transmembrane proteins that can serve as entry factors for enveloped viruses. Previous studies used pseudoviruses to delineate the role of TIM-1 and Axl in CHIKV entry. Conversely, here, we use the authentic CHIKV and cells ectopically expressing TIM-1 or Axl and demonstrate a role for TIM-1 in CHIKV infection. To further characterize TIM-1-dependent CHIKV infection, we generated cells expressing domain mutants of TIM-1. We show that point mutations in the phosphatidylserine binding site of TIM-1 lead to reduced cell binding, entry, and infection of CHIKV. Ectopic expression of TIM-1 renders immortalized keratinocytes permissive to CHIKV, whereas silencing of endogenously expressed TIM-1 in human hepatoma cells reduces CHIKV infection. Altogether, our findings indicate that, unlike Axl, TIM-1 readily promotes the productive entry of authentic CHIKV into target cells.
Collapse
Affiliation(s)
- Jared Kirui
- Centre for Experimental and Clinical Infection Research, TWINCORE, Institute for Experimental Virology, a Joint Venture between the Medical School Hannover and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany; (J.K.); (A.L.); (L.L.)
- Department of Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Yara Abidine
- Department of Clinical Microbiology, Umeå University, 90185 Umeå, Sweden; (Y.A.); (K.I.); (Y.-D.G.); (M.E.); (M.B.)
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, 90185 Umeå, Sweden
| | - Annasara Lenman
- Centre for Experimental and Clinical Infection Research, TWINCORE, Institute for Experimental Virology, a Joint Venture between the Medical School Hannover and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany; (J.K.); (A.L.); (L.L.)
- Department of Clinical Microbiology, Umeå University, 90185 Umeå, Sweden; (Y.A.); (K.I.); (Y.-D.G.); (M.E.); (M.B.)
| | - Koushikul Islam
- Department of Clinical Microbiology, Umeå University, 90185 Umeå, Sweden; (Y.A.); (K.I.); (Y.-D.G.); (M.E.); (M.B.)
| | - Yong-Dae Gwon
- Department of Clinical Microbiology, Umeå University, 90185 Umeå, Sweden; (Y.A.); (K.I.); (Y.-D.G.); (M.E.); (M.B.)
| | - Lisa Lasswitz
- Centre for Experimental and Clinical Infection Research, TWINCORE, Institute for Experimental Virology, a Joint Venture between the Medical School Hannover and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany; (J.K.); (A.L.); (L.L.)
- Department of Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Magnus Evander
- Department of Clinical Microbiology, Umeå University, 90185 Umeå, Sweden; (Y.A.); (K.I.); (Y.-D.G.); (M.E.); (M.B.)
| | - Marta Bally
- Department of Clinical Microbiology, Umeå University, 90185 Umeå, Sweden; (Y.A.); (K.I.); (Y.-D.G.); (M.E.); (M.B.)
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, 90185 Umeå, Sweden
| | - Gisa Gerold
- Centre for Experimental and Clinical Infection Research, TWINCORE, Institute for Experimental Virology, a Joint Venture between the Medical School Hannover and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany; (J.K.); (A.L.); (L.L.)
- Department of Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany
- Department of Clinical Microbiology, Umeå University, 90185 Umeå, Sweden; (Y.A.); (K.I.); (Y.-D.G.); (M.E.); (M.B.)
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, 90185 Umeå, Sweden
- Correspondence:
| |
Collapse
|
15
|
Xiao Z, Kandel A, Li L. Synergistic Activation of Bovine CD4+ T Cells by Neutrophils and IL-12. Pathogens 2021; 10:pathogens10060694. [PMID: 34204973 PMCID: PMC8228106 DOI: 10.3390/pathogens10060694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 05/27/2021] [Accepted: 05/29/2021] [Indexed: 12/12/2022] Open
Abstract
CD4+ T cell activation requires inflammatory cytokines to provide a third signal (3SI), such as interleukin-12 (IL-12). We recently reported that bovine neutrophils can enhance the activation of bovine CD4+ T cells. To explore the interactions between neutrophils and third signal cytokines in bovine CD4+ T cell activation, naïve CD4+ T cells were isolated from cattle lymph nodes and stimulated for 3.5 days with anti-bovine CD3 (first signal; 1SI), anti-bovine CD28 (second signal; 2SI), and recombinant human IL-12 (3SI) in the presence or absence of neutrophils harvested from the same animals. Indeed, the strongest activation was achieved in the presence of all three signals, as demonstrated by CD25 upregulation, IFNγ production in CD4+ T cells, and secretion of IFNγ and IL-2 in cell supernatants. More importantly, 1SI plus neutrophils led to enhanced CD25 expression that was further increased by IL-12, suggesting synergistic action by IL-12 and neutrophils. Consistently, neutrophils significantly increased IFNγ production in 1SI plus IL-12-stimulated CD4+ T cells. Our data suggest the synergy of neutrophils and IL-12 as a novel regulator on bovine CD4+ T cell activation in addition to three signals. This knowledge could assist the development of immune interventions for the control of infectious diseases in cattle.
Collapse
|
16
|
Significance of TIM-3 expression by CD4 + and CD8 + T lymphocytes in tumor-draining lymph nodes from patients with breast cancer. Mol Immunol 2020; 128:47-54. [PMID: 33068833 DOI: 10.1016/j.molimm.2020.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/23/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023]
Abstract
T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3), which is expressed by immune and nonimmune cells, has been shown to play immunoregulatory roles in the tumor microenvironment. In this study we assessed the expression of TIM-3 by T cells from tumor draining lymph nodes (TDLNs) of patients with breast cancer and its association with disease progression. Lymphocytes were isolated from 41 TDLNs, and flow cytometry was used to determine the expression of TIM-3 on CD4+ and CD8+ T cells, along with the simultaneous expression of CD25, Foxp3 and TIM-3 in CD4+ T cells. The results showed that the frequency of TIM-3+CD8+ T cells was associated with higher tumor grade, and the geometric mean fluorescence intensity (gMFI) of TIM-3 in CD4+ and CD8+ T cells was significantly higher in patients with more than 9 involved lymph nodes than those with fewer involved nodes. The gMFI of TIM3 in CD4+ T cells also showed a direct correlation with the number of metastatic lymph nodes. Phenotypic characterization of TIM-3+CD4+ T cells showed that the majority of CD4+TIM3+ lymphocytes were Foxp3 ̶ CD25 ̶, and the majority of Foxp3+CD25+ regulatory T cells were TIM-3-. Our findings showed that TIM-3 was expressed by CD4+, CD8+ and regulatory T cells in breast TDLNs, and that expression on CD4+ and CD8+ T cells was mostly associated with poor prognosticators such as a higher number of involved lymph nodes or higher tumor grade. More studies are required to confirm TIM-3 as a prognostic marker and a target for immunotherapy in breast cancer.
Collapse
|
17
|
Geng Q, Rohondia SO, Khan HJ, Jiao P, Dou QP. Small molecules as antagonists of co-inhibitory pathways for cancer immunotherapy: a patent review (2018-2019). Expert Opin Ther Pat 2020; 30:677-694. [PMID: 32715813 DOI: 10.1080/13543776.2020.1801640] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Therapeutic antibodies blocking co-inhibitory pathways do not attack tumor cells directly, but instead bind to their targeted proteins and mobilize the immune system to eradicate tumors. However, only a small fraction of patients with certain cancer types can benefit from the antibodies. Additionally, antibodies have shown serious immune-related adverse events in certain patients. Small-molecule antagonists may be a complementary and potentially synergistic approach to antibodies for patients with various cancers. AREAS COVERED The authors review the small molecules as antagonists of co-inhibitory pathway proteins, summarize their preliminary SARs, discuss biochemistry assays used in patents for the development of small molecules as novel antagonists. EXPERT OPINION The disclosed pharmacophores of small molecules as co-inhibitory pathway antagonists are represented by biphenyl derivatives, biaryl derivatives, teraryl derivatives, quateraryl derivatives, and oxadiazole/thiadiazole derivatives. However, these antagonists are still inferior to therapeutic antibodies in their inhibitory activities due to relatively flat of human co-inhibitory pathways proteins. Allosteric modulators may be an alternative approach. The more safety and efficacy evaluation trials of small-molecule antagonists targeting co-inhibitory pathways should be performed to demonstrate the proof-of-principle that small-molecule antagonists can result in sustained safety and antitumor response in the near future.
Collapse
Affiliation(s)
- Qiaohong Geng
- Department of Chemistry, Qilu Normal University , Jinan, China
| | - Sagar O Rohondia
- Barbara Ann Karmanos Cancer Institute, and Departments of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University , Detroit, MI, USA
| | - Harras J Khan
- Barbara Ann Karmanos Cancer Institute, and Departments of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University , Detroit, MI, USA
| | - Peifu Jiao
- Department of Chemistry, Qilu Normal University , Jinan, China
| | - Q Ping Dou
- Barbara Ann Karmanos Cancer Institute, and Departments of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University , Detroit, MI, USA
| |
Collapse
|
18
|
Huang P, Lv C, Zhang C, Feng H, Li C, Zhang H, Zhao X, Li M. Expression and significance of T-cell immunoglobulin mucin molecule 3 and its ligand galectin-9 in patients with adenomyosis. Gynecol Endocrinol 2020; 36:605-610. [PMID: 32319832 DOI: 10.1080/09513590.2020.1754788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
The T cell immunoglobulin mucin molecule 3 (TIM-3), as a negative regulatory immune checkpoint, plays an important role in cellular immunity by binding to its ligand galectin-9 (Gal-9). Under abnormal conditions, this pathway can regulate the depletion of T cells and suppress the immune response. Abnormal expression of TIM-3 and Gal-9 has been confirmed in a variety of cancers, recurrent miscarriages, chronic infectious diseases and autoimmune diseases. More and more studies have shown that immune factors play an important role in the pathogenesis of adenomyosis. However, few studies have attempted to explore their expression in adenomyosis. The ectopic and eutopic endometrium were collected from 15 women with adenomyosis and 13 women without adenomyosis. TIM-3/Gal-9 expression in endometrial tissue was detected using immunohistochemistry, western blot analysis and real-time PCR. We observed TIM-3/Gal-9 expression in both eutopic and ectopic endometria, with elevated expression in adenomyosis. These data suggest that TIM-3/Gal-9 may be involved in the development and progression of adenomyosis.
Collapse
Affiliation(s)
- Pu Huang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Chunzi Lv
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Cheng Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Hairong Feng
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Chunyan Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Hui Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Xingbo Zhao
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Mingjiang Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| |
Collapse
|
19
|
Shen H, Nibona E, Xu G, Al Hafiz MA, Ke X, Liang X, Yao Q, Zhong X, Zhou Q, Zhao H. Identification, expression pattern, and immune response of Tim-1 and Tim-4 in embryos and adult medaka (Oryzias latipes). JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2020; 334:235-244. [PMID: 32150339 DOI: 10.1002/jez.b.22939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 02/22/2020] [Accepted: 02/25/2020] [Indexed: 11/09/2022]
Abstract
T-cell immunoglobulin (Ig) and mucin domain-containing 1 (Tim-1) and Tim-4 are two members of the Tim family. In mammals, Tim-1 and Tim-4 are proteins mainly expressed in immune cells and are associated with immune response. In the present study, medaka Oryzias latipes' Tim-1 (OlTim-1) and OlTim-4 were identified and characterized using bioinformatics analyses. With the use of reverse-transcription polymerase chain reaction, the expression profiles of OlTim-1 and OlTim-4 were examined in embryos and adult fish and in immune tissues following the intraperitoneal injection of stimulants. The results revealed that OlTim-1 possesses a cytoplasmic region, a transmembrane region, a mucin domain, and an Ig-like domain, while OlTim-4 is composed of two Ig-like domains and a mucin domain, but without the transmembrane region and cytoplasmic region. OlTim-1 and OlTim-4 expressions are detectable from the gastrula stage on, indicating that they are zygotic genes. Furthermore, OlTim-1 and OlTim-4 are expressed ubiquitously in the adult. Administration of immune stimulants, namely lipopolysaccharides and polyinosinic:polycytidylic acid, significantly increased the expression levels of OlTim-1 and OlTim-4 in the liver and intestine within 1 day and in the head, kidney, and spleen within 3 to 4 days postinjection. These results suggest that OlTim-1 and OlTim-4 are possibly involved in both innate and adaptive immunities.
Collapse
Affiliation(s)
- Hao Shen
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, Hubei, China
| | - Emile Nibona
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, Hubei, China
| | - Gongyu Xu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, Hubei, China
| | - Md Abdullah Al Hafiz
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, Hubei, China
| | - Xiaomei Ke
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, Hubei, China
| | - Xiaoting Liang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, Hubei, China
| | - Qiting Yao
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, Hubei, China
| | - Xueping Zhong
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, Hubei, China
| | - Qingchun Zhou
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, Hubei, China
| | - Haobin Zhao
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, Hubei, China
| |
Collapse
|
20
|
Huang R, Jing X, Huang X, Pan Y, Fang Y, Liang G, Liao Z, Wang H, Chen Z, Zhang Y. Bifunctional Naphthoquinone Aromatic Amide-Oxime Derivatives Exert Combined Immunotherapeutic and Antitumor Effects through Simultaneous Targeting of Indoleamine-2,3-dioxygenase and Signal Transducer and Activator of Transcription 3. J Med Chem 2020; 63:1544-1563. [PMID: 31999451 DOI: 10.1021/acs.jmedchem.9b01386] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Indoleamine-2,3-dioxygenase 1 (IDO1) and signal transducer and activator of transcription 3 (STAT3) are important targets in the tumor microenvironment for cancer therapy. In the present study, a set of naphthoquinone aromatic amide-oxime derivatives were designed, which stimulated the immune response via IDO1 inhibition and simultaneously displayed powerful antitumor activity against three selected cancer cell lines through suppressing STAT3 signaling. The representative compound 8u bound effectively to IDO1, with greater inhibitory activity relative to the commercial IDO1 inhibitor 4-amino-N-(3-chloro-4-fluorophenyl)-N'-hydroxy-1,2,5-oxadiazole-3-carboximidamide (IDO5L) in addition to the efficient suppression of nuclear translocation of STAT3. Consistently, in vivo assays demonstrated a higher antiproliferative activity of compound 8u in both wild-type B16-F10 isograft tumors and an athymic HepG2 xenograft model relative to 1-methyl-l-tryptophan (1-MT) and doxorubicin (DOX). This bifunctional compound with dual immunotherapeutic and anticancer efficacy may represent a new generation of highly efficacious drug candidates for cancer therapy.
Collapse
Affiliation(s)
- Rizhen Huang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China) , School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University , Guilin 541004 , China.,School of Pharmacy , Guilin Medical University , Guilin 541004 , China.,Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research and School of Chemistry and Chemical Engineering , Southeast University , Nanjing 211189 , China
| | - Xiaoteng Jing
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China) , School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University , Guilin 541004 , China
| | - Xiaochao Huang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research and School of Chemistry and Chemical Engineering , Southeast University , Nanjing 211189 , China
| | - Yingming Pan
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China) , School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University , Guilin 541004 , China
| | - Yilin Fang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China) , School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University , Guilin 541004 , China
| | - Guibin Liang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China) , School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University , Guilin 541004 , China
| | - Zhixin Liao
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research and School of Chemistry and Chemical Engineering , Southeast University , Nanjing 211189 , China
| | - Hengshan Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China) , School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University , Guilin 541004 , China
| | - Zhenfeng Chen
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China) , School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University , Guilin 541004 , China
| | - Ye Zhang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China) , School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University , Guilin 541004 , China.,School of Pharmacy , Guilin Medical University , Guilin 541004 , China
| |
Collapse
|
21
|
Jeong S, Park SH. Co-Stimulatory Receptors in Cancers and Their Implications for Cancer Immunotherapy. Immune Netw 2020; 20:e3. [PMID: 32158591 PMCID: PMC7049585 DOI: 10.4110/in.2020.20.e3] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 01/27/2020] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs), including anti-PD-1 and anti-CTLA-4 therapeutic agents, are now approved by the Food and Drug Administration for treatment of various types of cancer. However, the therapeutic efficacy of ICIs varies among patients and cancer types. Moreover, most patients do not develop durable antitumor responses after ICI therapy due to an ephemeral reversal of T-cell dysfunction. As co-stimulatory receptors play key roles in regulating the effector functions of T cells, activating co-stimulatory pathways may improve checkpoint inhibition efficacy, and lead to durable antitumor responses. Here, we review recent advances in our understating of co-stimulatory receptors in cancers, providing the necessary groundwork for the rational design of cancer immunotherapy.
Collapse
Affiliation(s)
- Seongju Jeong
- Biomedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon 34141, Korea
| | - Su-Hyung Park
- Biomedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon 34141, Korea.,Laboratory of Translational Immunology and Vaccinology, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Korea
| |
Collapse
|
22
|
Abstract
The role of inflammation in cardiovascular disease (CVD) is now widely accepted. Immune cells, including T cells, are influenced by inflammatory signals and contribute to the onset and progression of CVD. T cell activation is modulated by T cell co-stimulation and co-inhibition pathways. Immune checkpoint inhibitors (ICIs) targeting T cell inhibition pathways have revolutionized cancer treatment and improved survival in patients with cancer. However, ICIs might induce cardiovascular toxicity via T cell re-invigoration. With the rising use of ICIs for cancer treatment, a timely overview of the role of T cell co-stimulation and inhibition molecules in CVD is desirable. In this Review, the importance of these molecules in the pathogenesis of CVD is highlighted in preclinical studies on models of CVD such as vein graft disease, myocarditis, graft arterial disease, post-ischaemic neovascularization and atherosclerosis. This Review also discusses the therapeutic potential of targeting T cell co-stimulation and inhibition pathways to treat CVD, as well as the possible cardiovascular benefits and adverse events after treatment. Finally, the Review emphasizes that patients with cancer who are treated with ICIs should be monitored for CVD given the reported association between the use of ICIs and the risk of cardiovascular toxicity.
Collapse
|
23
|
|
24
|
Yeung MY, Grimmig T, Sayegh MH. Costimulation Blockade in Transplantation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1189:267-312. [PMID: 31758538 DOI: 10.1007/978-981-32-9717-3_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
T cells play a pivotal role in orchestrating immune responses directed against a foreign (allogeneic) graft. For T cells to become fully activated, the T-cell receptor (TCR) must interact with the major histocompatibility complex (MHC) plus peptide complex on antigen-presenting cells (APCs), followed by a second "positive" costimulatory signal. In the absence of this second signal, T cells become anergic or undergo deletion. By blocking positive costimulatory signaling, T-cell allo-responses can be aborted, thus preventing graft rejection and promoting long-term allograft survival and possibly tolerance (Alegre ML, Najafian N, Curr Mol Med 6:843-857, 2006; Li XC, Rothstein DM, Sayegh MH, Immunol Rev 229:271-293, 2009). In addition, costimulatory molecules can provide negative "coinhibitory" signals that inhibit T-cell activation and terminate immune responses; strategies to promote these pathways can also lead to graft tolerance (Boenisch O, Sayegh MH, Najafian N, Curr Opin Organ Transplant 13:373-378, 2008). However, T-cell costimulation involves an incredibly complex array of interactions that may act simultaneously or at different times in the immune response and whose relative importance varies depending on the different T-cell subsets and activation status. In transplantation, the presence of foreign alloantigen incites not only destructive T effector cells but also protective regulatory T cells, the balance of which ultimately determines the fate of the allograft (Lechler RI, Garden OA, Turka LA, Nat Rev Immunol 3:147-158, 2003). Since the processes of alloantigen-specific rejection and regulation both require activation of T cells, costimulatory interactions may have opposing or synergistic roles depending on the cell being targeted. Such complexities present both challenges and opportunities in targeting T-cell costimulatory pathways for therapeutic purposes. In this chapter, we summarize our current knowledge of the various costimulatory pathways in transplantation and review the current state and challenges of harnessing these pathways to promote graft tolerance (summarized in Table 10.1).
Collapse
Affiliation(s)
- Melissa Y Yeung
- Department of Medicine, Renal Division, Brigham and Women's Hospital, Boston, MA, USA. .,Harvard Medical School, Boston, MA, USA.
| | - Tanja Grimmig
- Department of Surgery, Molecular Oncology and Immunology, University of Wuerzburg, Wuerzburg, Germany
| | - Mohamed H Sayegh
- Department of Medicine, Renal Division, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Department of Medicine and Immunology, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
25
|
Malaker SA, Pedram K, Ferracane MJ, Bensing BA, Krishnan V, Pett C, Yu J, Woods EC, Kramer JR, Westerlind U, Dorigo O, Bertozzi CR. The mucin-selective protease StcE enables molecular and functional analysis of human cancer-associated mucins. Proc Natl Acad Sci U S A 2019; 116:7278-7287. [PMID: 30910957 PMCID: PMC6462054 DOI: 10.1073/pnas.1813020116] [Citation(s) in RCA: 191] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mucin domains are densely O-glycosylated modular protein domains that are found in a wide variety of cell surface and secreted proteins. Mucin-domain glycoproteins are known to be key players in a host of human diseases, especially cancer, wherein mucin expression and glycosylation patterns are altered. Mucin biology has been difficult to study at the molecular level, in part, because methods to manipulate and structurally characterize mucin domains are lacking. Here, we demonstrate that secreted protease of C1 esterase inhibitor (StcE), a bacterial protease from Escherichia coli, cleaves mucin domains by recognizing a discrete peptide- and glycan-based motif. We exploited StcE's unique properties to improve sequence coverage, glycosite mapping, and glycoform analysis of recombinant human mucins by mass spectrometry. We also found that StcE digests cancer-associated mucins from cultured cells and from ascites fluid derived from patients with ovarian cancer. Finally, using StcE, we discovered that sialic acid-binding Ig-type lectin-7 (Siglec-7), a glycoimmune checkpoint receptor, selectively binds sialomucins as biological ligands, whereas the related receptor Siglec-9 does not. Mucin-selective proteolysis, as exemplified by StcE, is therefore a powerful tool for the study of mucin domain structure and function.
Collapse
Affiliation(s)
- Stacy A Malaker
- Department of Chemistry, Stanford University, Stanford, CA 94305
| | - Kayvon Pedram
- Department of Chemistry, Stanford University, Stanford, CA 94305
| | | | - Barbara A Bensing
- Department of Medicine, San Francisco Veterans Affairs Medical Center and University of California, San Francisco, CA 94143
| | - Venkatesh Krishnan
- Stanford Women's Cancer Center, Division of Gynecologic Oncology, Stanford University, Stanford, CA 94305
| | - Christian Pett
- Leibniz-Institut für Analytische Wissenschaften (ISAS), 44227 Dortmund, Germany
- Department of Chemistry, Umeå University, 901 87 Umeå, Sweden
| | - Jin Yu
- Leibniz-Institut für Analytische Wissenschaften (ISAS), 44227 Dortmund, Germany
| | - Elliot C Woods
- Department of Chemistry, Stanford University, Stanford, CA 94305
| | - Jessica R Kramer
- Department of Bioengineering, University of Utah, Salt Lake City, UT 84112
| | - Ulrika Westerlind
- Leibniz-Institut für Analytische Wissenschaften (ISAS), 44227 Dortmund, Germany
- Department of Chemistry, Umeå University, 901 87 Umeå, Sweden
| | - Oliver Dorigo
- Stanford Women's Cancer Center, Division of Gynecologic Oncology, Stanford University, Stanford, CA 94305
| | - Carolyn R Bertozzi
- Department of Chemistry, Stanford University, Stanford, CA 94305;
- Howard Hughes Medical Institute, Stanford, CA 94305
| |
Collapse
|
26
|
Song J, Yu J, Prayogo GW, Cao W, Wu Y, Jia Z, Zhang A. Understanding kidney injury molecule 1: a novel immune factor in kidney pathophysiology. Am J Transl Res 2019; 11:1219-1229. [PMID: 30972157 PMCID: PMC6456506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 01/02/2019] [Indexed: 06/09/2023]
Abstract
Kidney injury molecule 1 (KIM-1) is a type I membrane protein, comprising an extracellular portion and a cytoplasmic portion. It is also named as HAVCR1 (Hepatitis A virus cellular receptor 1) or TIM1 (T-cell immunoglobulin mucin receptor 1), and is expressed in the kidney, liver, and spleen. KIM-1 plays different roles via various molecular targets in immune diseases and kidney injury. KIM-1 is involved in HAV infections, autoimmunity, immune tolerance, and atopic diseases. The urinary KIM-1 level is closely related to its tissue level, and correspondingly related to kidney tissue damage. KIM-1 is not only an early biomarker of acute kidney injury (AKI), but also has a potential role in predicting the long-term renal outcome. In this review, we provide a summary of KIM-1's activities, focusing on the latest studies concerning the important roles of KIM-1 in the immune system and kidney diseases.
Collapse
Affiliation(s)
- Jiayu Song
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical UniversityNanjing 210029, China
| | - Jing Yu
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical UniversityNanjing 210029, China
| | - Gabriella Wenda Prayogo
- Department of Endocrinology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
| | - Weidong Cao
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical UniversityNanjing 210029, China
| | - Yimei Wu
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
| | - Zhanjun Jia
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical UniversityNanjing 210029, China
| | - Aihua Zhang
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical UniversityNanjing 210029, China
| |
Collapse
|
27
|
Zhao M, Guo W, Wu Y, Yang C, Zhong L, Deng G, Zhu Y, Liu W, Gu Y, Lu Y, Kong L, Meng X, Xu Q, Sun Y. SHP2 inhibition triggers anti-tumor immunity and synergizes with PD-1 blockade. Acta Pharm Sin B 2019; 9:304-315. [PMID: 30972278 PMCID: PMC6437555 DOI: 10.1016/j.apsb.2018.08.009] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/18/2018] [Accepted: 08/30/2018] [Indexed: 12/21/2022] Open
Abstract
Tyrosine phosphatase SHP2 is a promising drug target in cancer immunotherapy due to its bidirectional role in both tumor growth promotion and T-cell inactivation. Its allosteric inhibitor SHP099 is known to inhibit cancer cell growth both in vitro and in vivo. However, whether SHP099-mediated SHP2 inhibition retards tumor growth in vivo via anti-tumor immunity remains elusive. To address this, a CT-26 colon cancer xenograft model was established in mice since this cell line is insensitive to SHP099. Consequently, SHP099 minimally affected CT-26 tumor growth in immuno-deficient nude mice, but significantly decreased the tumor burden in CT-26 tumor-bearing mice with intact immune system. SHP099 augmented anti-tumor immunity, as shown by the elevated proportion of CD8+IFN-γ+ T cells and the upregulation of cytotoxic T-cell related genes including Granzyme B andPerforin, which decreased the tumor load. In addition, tumor growth in mice with SHP2-deficient T-cells was markedly slowed down because of enhanced anti-tumor responses. Finally, the combination of SHP099 and anti-PD-1 antibody showed a higher therapeutic efficacy than either monotherapy in controlling tumor growth in two colon cancer xenograft models, indicating that these agents complement each other. Our study suggests that SHP2 inhibitor SHP099 is a promising candidate drug for cancer immunotherapy.
Collapse
|
28
|
Hua S, Chen F, Wang X, Wang Y, Gou S. Pt(IV) hybrids containing a TDO inhibitor serve as potential anticancer immunomodulators. J Inorg Biochem 2019; 195:130-140. [PMID: 30952082 DOI: 10.1016/j.jinorgbio.2019.02.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 02/07/2019] [Accepted: 02/08/2019] [Indexed: 12/21/2022]
Abstract
Tryptophan 2,3-dioxygenase (TDO), an immunosuppressive enzyme, can involve in immune evasion and tumor tolerance. TDO inhibitors can boost the efficacy of chemotherapeutics by promoting immunity. Herein, a strategy to introduce a TDO inhibitor into Pt(IV) complexes for reversing tumor immune suppression was adopted. A mono-modified Pt(IV) complex, 3, displayed significant antitumor activity against human liver cancer cells. Flow cytometry study revealed that complex 3 could induce cell death via a mitochondrial-dependent apoptosis pathway and arrest the cell cycle at S phase. Furthermore, complex 3 was effective to enhance T-cell immune responses by inhibiting the TDO enzyme expression to block the kynurenine production and inactivating the downstream of aryl hydrocarbon receptor (AHR).
Collapse
Affiliation(s)
- Shixian Hua
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Feihong Chen
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China; Jiangsu Province Hi-Tech Key Laboratory for Bio-medical Research, Southeast University, Nanjing 211189, China
| | - Xinyi Wang
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Yuanjiang Wang
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Shaohua Gou
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China; Jiangsu Province Hi-Tech Key Laboratory for Bio-medical Research, Southeast University, Nanjing 211189, China.
| |
Collapse
|
29
|
Kumar P, Bhattacharya P, Prabhakar BS. A comprehensive review on the role of co-signaling receptors and Treg homeostasis in autoimmunity and tumor immunity. J Autoimmun 2018; 95:77-99. [PMID: 30174217 PMCID: PMC6289740 DOI: 10.1016/j.jaut.2018.08.007] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 08/22/2018] [Accepted: 08/26/2018] [Indexed: 12/22/2022]
Abstract
The immune system ensures optimum T-effector (Teff) immune responses against invading microbes and tumor antigens while preventing inappropriate autoimmune responses against self-antigens with the help of T-regulatory (Treg) cells. Thus, Treg and Teff cells help maintain immune homeostasis through mutual regulation. While Tregs can contribute to tumor immune evasion by suppressing anti-tumor Teff response, loss of Treg function can result in Teff responses against self-antigens leading to autoimmune disease. Thus, loss of homeostatic balance between Teff/Treg cells is often associated with both cancer and autoimmunity. Co-stimulatory and co-inhibitory receptors, collectively known as co-signaling receptors, play an indispensable role in the regulation of Teff and Treg cell expansion and function and thus play critical roles in modulating autoimmune and anti-tumor immune responses. Over the past three decades, considerable efforts have been made to understand the biology of co-signaling receptors and their role in immune homeostasis. Mutations in co-inhibitory receptors such as CTLA4 and PD1 are associated with Treg dysfunction, and autoimmune diseases in mice and humans. On the other hand, growing tumors evade immune surveillance by exploiting co-inhibitory signaling through expression of CTLA4, PD1 and PDL-1. Immune checkpoint blockade (ICB) using anti-CTLA4 and anti-PD1 has drawn considerable attention towards co-signaling receptors in tumor immunology and created renewed interest in studying other co-signaling receptors, which until recently have not been as well studied. In addition to co-inhibitory receptors, co-stimulatory receptors like OX40, GITR and 4-1BB have also been widely implicated in immune homeostasis and T-cell stimulation, and use of agonistic antibodies against OX40, GITR and 4-1BB has been effective in causing tumor regression. Although ICB has seen unprecedented success in cancer treatment, autoimmune adverse events arising from ICB due to loss of Treg homeostasis poses a major obstacle. Herein, we comprehensively review the role of various co-stimulatory and co-inhibitory receptors in Treg biology and immune homeostasis, autoimmunity, and anti-tumor immunity. Furthermore, we discuss the autoimmune adverse events arising upon targeting these co-signaling receptors to augment anti-tumor immune responses.
Collapse
Affiliation(s)
- Prabhakaran Kumar
- Department of Microbiology and Immunology, University of Illinois-College of Medicine, Chicago, IL, USA
| | - Palash Bhattacharya
- Department of Microbiology and Immunology, University of Illinois-College of Medicine, Chicago, IL, USA
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois-College of Medicine, Chicago, IL, USA; Department of Ophthalmology, Associate Dean for Technological Innovation and Training, University of Illinois College of Medicine, Room E-705, (M/C 790), 835 S. Wolcott Ave, Chicago, IL, 60612, USA.
| |
Collapse
|
30
|
Paul B, Kang S, Zheng Z, Kang Y. The challenges of checkpoint inhibition in the treatment of multiple myeloma. Cell Immunol 2018; 334:87-98. [PMID: 30342750 DOI: 10.1016/j.cellimm.2018.10.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 10/12/2018] [Indexed: 12/22/2022]
Abstract
Despite significant improvements in the overall survival of patients with multiple myeloma (MM) over the past 15 years, the disease remains incurable. Treatment options are limited for patients who have relapsed or are refractory to immunomodulatory drugs (IMiDs), proteasome inhibitors, and monoclonal antibodies. In these patients, immunotherapies such as checkpoint inhibitors, oncolytic vaccines, and chimeric antigen receptor (CAR) T cells provide a potentially effective alternative treatment. While checkpoint inhibitors are effective in prolonging overall survival in some patients with advanced solid cancers and Hodgkin lymphoma, they have not demonstrated significant activity as a single agent in MM. In fact the combination of checkpoint inhibitors with IMiDs was recently found to increase the risk of death in myeloma patients. These challenges highlight the need for a better understanding of immune dysregulation in myeloma patients, and the mechanisms of action of- and resistance to- checkpoint inhibitors. In this review, we summarize immune dysfunction in patients with MM, and review the preclinical and clinical data regarding checkpoint inhibitors in myeloma. We conclude by proposing strategies to improve the efficacy and safety of checkpoint inhibitors in this population.
Collapse
Affiliation(s)
- Barry Paul
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC 27710, USA
| | - Shuqi Kang
- University of Illinois College of Medicine at Peoria, Peoria, IL 61605, USA
| | - Zhihong Zheng
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
| | - Yubin Kang
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
31
|
Li X, Liu Y, Tian H. Current Developments in Pt(IV) Prodrugs Conjugated with Bioactive Ligands. Bioinorg Chem Appl 2018; 2018:8276139. [PMID: 30402082 PMCID: PMC6191961 DOI: 10.1155/2018/8276139] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 08/19/2018] [Accepted: 09/06/2018] [Indexed: 12/21/2022] Open
Abstract
To overcome the side effects of and resistance to cisplatin, a variety of Pt(IV) prodrugs were designed and synthesized via different modifications including combination with lipid chains to increase hydrophobicity, conjugation with short peptide chains or nanoparticles to improve drug delivery, or addition of bioactive ligands to the axial positions of Pt(IV) complexes to exert dual-function effects. This review summarizes the recent progress in the development of Pt(IV) prodrugs conjugated with bioactive-targeting ligands, including histone deacetylase inhibitors, p53 agonists, alkylating agents, and nonsteroidal anti-inflammatory agents. Although Pt(IV) complexes that conjugated with bioactive ligands show satisfactory anticancer effects, none has been approved for clinical use. Therefore, we hope that this review will contribute to further study and development of Pt(IV) complexes conjugated with bioactive and other ligands.
Collapse
Affiliation(s)
- Xuejiao Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Yahong Liu
- Tianjin Binjiang Pharma, Inc., Tianjin 300192, China
| | - Hongqi Tian
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| |
Collapse
|
32
|
Gorman JV, Colgan JD. Acute stimulation generates Tim-3-expressing T helper type 1 CD4 T cells that persist in vivo and show enhanced effector function. Immunology 2018; 154:418-433. [PMID: 29315553 DOI: 10.1111/imm.12890] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 12/14/2017] [Accepted: 12/22/2017] [Indexed: 12/20/2022] Open
Abstract
T-cell immunoglobulin and mucin domain 3 (Tim-3) is a surface receptor expressed by T helper type 1 (Th1) effector CD4 T cells, which are critical for defence against intracellular pathogens and have been implicated in autoimmune disease. Previous studies showed that Tim-3 expression makes Th1 cells more susceptible to apoptosis and also marks functionally impaired T cells that arise due to chronic stimulation. However, other studies suggested that Tim-3-expressing Th1 cells do not always have these properties. To further define the relationship between Tim-3 and Th1 cell function, we analysed the characteristics of Th1 cells that expressed Tim-3 in response to brief stimulation in vitro or an acute viral infection in vivo. As expected, cultured CD4 T cells began expressing Tim-3 during Th1 differentiation and secondary stimulation generated Tim-3- and Tim-3+ fractions that were separated and further analysed. When injected into naive mice, Tim-3+ cells down-regulated Tim-3 and survived equally well compared with Tim-3- cells. Further, Tim-3- and Tim-3+ Th1 cells had similar functional responses when transferred into naive mice that were subsequently infected with lymphocytic choriomeningitis virus (LCMV). Cultured Th1 cells that expressed Tim-3 following T-cell receptor stimulation had a greater capacity to express signature Th1 cytokines than their Tim-3- counterparts and showed differential expression of genes that regulate CD4 T-cell function. Consistent with these findings, Tim-3+ Th1 cells generated in response to LCMV infection displayed augmented effector function relative to Tim-3- cells. These results suggest that Tim-3 expression by Th1 cells responding to acute stimulation can mark cells that are functionally competent and have an augmented ability to produce cytokines.
Collapse
Affiliation(s)
- Jacob V Gorman
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA
| | - John D Colgan
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA.,Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
33
|
de Mingo Pulido Á, Gardner A, Hiebler S, Soliman H, Rugo HS, Krummel MF, Coussens LM, Ruffell B. TIM-3 Regulates CD103 + Dendritic Cell Function and Response to Chemotherapy in Breast Cancer. Cancer Cell 2018; 33:60-74.e6. [PMID: 29316433 PMCID: PMC5764109 DOI: 10.1016/j.ccell.2017.11.019] [Citation(s) in RCA: 278] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/13/2017] [Accepted: 11/29/2017] [Indexed: 12/18/2022]
Abstract
Intratumoral CD103+ dendritic cells (DCs) are necessary for anti-tumor immunity. Here we evaluated the expression of immune regulators by CD103+ DCs in a murine model of breast cancer and identified expression of TIM-3 as a target for therapy. Anti-TIM-3 antibody improved response to paclitaxel chemotherapy in models of triple-negative and luminal B disease, with no evidence of toxicity. Combined efficacy was CD8+ T cell dependent and associated with increased granzyme B expression; however, TIM-3 expression was predominantly localized to myeloid cells in both human and murine tumors. Gene expression analysis identified upregulation of Cxcl9 within intratumoral DCs during combination therapy, and therapeutic efficacy was ablated by CXCR3 blockade, Batf3 deficiency, or Irf8 deficiency.
Collapse
Affiliation(s)
- Álvaro de Mingo Pulido
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive SRB-2, Tampa, FL 33612, USA
| | - Alycia Gardner
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive SRB-2, Tampa, FL 33612, USA; Cancer Biology PhD Program, University of South Florida, Tampa, FL 33620, USA
| | - Shandi Hiebler
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive SRB-2, Tampa, FL 33612, USA
| | - Hatem Soliman
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive SRB-2, Tampa, FL 33612, USA; Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Hope S Rugo
- Department of Medicine and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94143, USA
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, CA 94143, USA
| | - Lisa M Coussens
- Department of Cell, Developmental & Cancer Biology, and Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Brian Ruffell
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive SRB-2, Tampa, FL 33612, USA; Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA.
| |
Collapse
|
34
|
Hollmann T, Kim TK, Tirloni L, Radulović ŽM, Pinto AFM, Diedrich JK, Yates JR, da Silva Vaz I, Mulenga A. Identification and characterization of proteins in the Amblyomma americanum tick cement cone. Int J Parasitol 2017; 48:211-224. [PMID: 29258831 DOI: 10.1016/j.ijpara.2017.08.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 08/26/2017] [Indexed: 01/08/2023]
Abstract
The adaptation of hard ticks to feed for long periods is facilitated by the cement cone, which securely anchors the tick mouthparts onto host skin and protects the tick from being groomed off by the host. Thus, preventing tick cement deposition is an attractive target for the development of innovative tick control. We used LC-MS/MS sequencing to identify 160 Amblyomma americanum tick cement proteins that include glycine-rich proteins (GRP, 19%), protease inhibitors (12%), proteins of unknown function (11%), mucin (4%), detoxification, storage, and lipocalin at 1% each, and housekeeping proteins (50%). Spatiotemporal transcription analysis showing mRNA expression in multiple tick organs and transcript abundance increasing with feeding suggest that selected GRPs (n = 13) regulate multiple tick feeding functions, being classified as constitutively expressed (CE), feeding induced (FI), and up-regulated with feeding (UR). We show that transcription of CE GRPs is likely under the control of tick appetence associated factors in that mRNA abundance increased several thousand fold in 1 week old adult ticks, the time period that coincides with tick attainment of appetence. Given the high number of targets, we synthesized and injected unfed ticks with combinatorial (co) double stranded (ds)RNA and disrupted GRP mRNA in clusters according to similar transcription patterns: CE (n = 3), FI, (n = 4), and UR (n = 6) to streamline the work. Our data suggest that CE and FI GRPs are important for maintenance of the tick feeding site in that reddening and subsequent bleeding were observed around the mouthparts of CE and FI GRP co-dsRNA injected ticks during feeding. Furthermore, although not significantly different, indices for blood meal size and fecundity were apparently reduced in FI and UR ticks. We discuss our data with reference to A. americanum tick feeding physiology.
Collapse
Affiliation(s)
- Taylor Hollmann
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA
| | - Tae Kwon Kim
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA
| | - Lucas Tirloni
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA; Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Željko M Radulović
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA
| | - Antônio F M Pinto
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil; Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA; Mass Spectrometry Core, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jolene K Diedrich
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA; Mass Spectrometry Core, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - John R Yates
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Itabajara da Silva Vaz
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Albert Mulenga
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
35
|
Pouladian M, Ganjalikhani-Hakemi M, Alsahebfosul F, Homayouni V, Khosravi S, Etemadifar M, Mazrouei F, Salehi R. The +4259A>C polymorphism of TIM-3 but not -1637C>T polymorphism of TIM-1 is associated with Multiple sclerosis in Isfahan population. Mult Scler Relat Disord 2017; 18:152-156. [PMID: 29141799 DOI: 10.1016/j.msard.2017.09.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 09/24/2017] [Accepted: 09/27/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS) which initiated and mediated by autoreactive T helper1 cells directed against myelin antigens. One of T cell surface receptors is T cell immunoglobulin and mucin domain (TIM) family which its importance in immunology is recently discovered. These molecules have important immunological function by regulation of T effector cells. METHODS In the present study, we analyzed the frequency of +4259A>C polymorphism in TIM-3 and -1637C>T polymorphism in TIM-1 gene in MS patients and healthy controls using restriction fragment length polymorphism-polymerase chain reaction (RFLP-PCR) method. RESULTS We found that the polymorphism +4259A>C in exon 3 of the TIM-3 gene is associated with susceptibility to the MS (P = 0.029, OR (95%CI) = 1.841) but the other polymorphism, -1637T>C, in the promoter region of TIM-1 is not (p= 0.064). CONCLUSION Our findings suggest that +4259A>C polymorphism in TIM-3 gene may be one of the important genetic factors associated with the MS susceptibility among Iranian populations.
Collapse
Affiliation(s)
- Masoumeh Pouladian
- Department of Immunology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | | | - Fereshteh Alsahebfosul
- Department of Immunology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Vida Homayouni
- Department of Immunology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Sharifeh Khosravi
- Department of Cellular & Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Masoud Etemadifar
- Department of Neurosciences, Al-Zahra Hospital, Isfahan University of Medical Science, Isfahan, Iran
| | - Fariba Mazrouei
- Department of Immunology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Rasoul Salehi
- Department of Cellular & Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| |
Collapse
|
36
|
Sims B, Farrow AL, Williams SD, Bansal A, Krendelchtchikov A, Gu L, Matthews QL. Role of TIM-4 in exosome-dependent entry of HIV-1 into human immune cells. Int J Nanomedicine 2017; 12:4823-4833. [PMID: 28740388 PMCID: PMC5505621 DOI: 10.2147/ijn.s132762] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Exosomes, 30–200 nm nanostructures secreted from donor cells and internalized by recipient cells, can play an important role in the cellular entry of some viruses. These microvesicles are actively secreted into various body fluids, including blood, urine, saliva, cerebrospinal fluid, and breast milk. We successfully isolated exosomes from human breast milk and plasma. The size and concentration of purified exosomes were measured by nanoparticle tracking, while Western blotting confirmed the presence of the exosomal-associated proteins CD9 and CD63, clathrin, and T cell immunoglobulin and mucin proteins (TIMs). Through viral infection assays, we determined that HIV-1 utilizes an exosome-dependent mechanism for entry into human immune cells. The virus contains high amounts of phosphatidylserine (PtdSer) and may bind PtdSer receptors, such as TIMs. This mechanism is supported by our findings that exosomes from multiple sources increased HIV-1 entry into T cells and macrophages, and viral entry was potently blocked with anti-TIM-4 antibodies.
Collapse
Affiliation(s)
- Brian Sims
- Division of Neonatology, Department of Pediatrics.,Department of Cell, Developmental and Integrative Biology.,Center for AIDS Research
| | | | - Sparkle D Williams
- Division of Neonatology, Department of Pediatrics.,Department of Cell, Developmental and Integrative Biology
| | | | - Alexandre Krendelchtchikov
- Division of Neonatology, Department of Pediatrics.,Department of Cell, Developmental and Integrative Biology.,Division of Infectious Diseases
| | - Linlin Gu
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham
| | - Qiana L Matthews
- Center for AIDS Research.,Division of Infectious Diseases.,Microbiology Program, Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL, USA
| |
Collapse
|
37
|
Schofield L, Ioannidis LJ, Karl S, Robinson LJ, Tan QY, Poole DP, Betuela I, Hill DL, Siba PM, Hansen DS, Mueller I, Eriksson EM. Synergistic effect of IL-12 and IL-18 induces TIM3 regulation of γδ T cell function and decreases the risk of clinical malaria in children living in Papua New Guinea. BMC Med 2017; 15:114. [PMID: 28615061 PMCID: PMC5471992 DOI: 10.1186/s12916-017-0883-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 05/22/2017] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND γδ T cells are important for both protective immunity and immunopathogenesis during malaria infection. However, the immunological processes determining beneficial or detrimental effects on disease outcome remain elusive. The aim of this study was to examine expression and regulatory effect of the inhibitory receptor T-cell immunoglobulin domain and mucin domain 3 (TIM3) on γδ T cells. While TIM3 expression and function on conventional αβ T cells have been clearly defined, the equivalent characterization on γδ T cells and associations with disease outcomes is limited. This study investigated the functional capacity of TIM3+ γδ T cells and the underlying mechanisms contributing to TIM3 upregulation and established an association with malaria disease outcomes. METHODS We analyzed TIM3 expression on γδ T cells in 132 children aged 5-10 years living in malaria endemic areas of Papua New Guinea. TIM3 upregulation and effector functions of TIM3+ γδ T cells were assessed following in vitro stimulation with parasite-infected erythrocytes, phosphoantigen and/or cytokines. Associations between the proportion of TIM3-expressing cells and the molecular force of infection were tested using negative binomial regression and in a Cox proportional hazards model for time to first clinical episode. Multivariable analyses to determine the association of TIM3 and IL-18 levels were conducted using general linear models. Malaria infection mouse models were utilized to experimentally investigate the relationship between repeated exposure and TIM3 upregulation. RESULTS This study demonstrates that even in the absence of an active malaria infection, children of malaria endemic areas have an atypical population of TIM3-expressing γδ T cells (mean frequency TIM3+ of total γδ T cells 15.2% ± 12). Crucial factors required for γδ T cell TIM3 upregulation include IL-12/IL-18, and plasma IL-18 was associated with TIM3 expression (P = 0.002). Additionally, we show a relationship between TIM3 expression and infection with distinct parasite clones during repeated exposure. TIM3+ γδ T cells were functionally impaired and were associated with asymptomatic malaria infection (hazard ratio 0.54, P = 0.032). CONCLUSIONS Collectively our data demonstrate a novel role for IL-12/IL-18 in shaping the innate immune response and provide fundamental insight into aspects of γδ T cell immunoregulation. Furthermore, we show that TIM3 represents an important γδ T cell regulatory component involved in minimizing malaria symptoms.
Collapse
Affiliation(s)
- Louis Schofield
- Walter and Eliza Hall Institute of Medical Research, Division of Population Health and Immunity, Melbourne, VIC, 3052, Australia.,Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, 4811, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Lisa J Ioannidis
- Walter and Eliza Hall Institute of Medical Research, Division of Population Health and Immunity, Melbourne, VIC, 3052, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Stephan Karl
- Walter and Eliza Hall Institute of Medical Research, Division of Population Health and Immunity, Melbourne, VIC, 3052, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Leanne J Robinson
- Walter and Eliza Hall Institute of Medical Research, Division of Population Health and Immunity, Melbourne, VIC, 3052, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, 3052, Australia.,Papua New Guinea Institute of Medical Research, Goroka and Madang, Papua New Guinea.,Burnet Institute, Melbourne, VIC, 3004, Australia
| | - Qiao Y Tan
- Walter and Eliza Hall Institute of Medical Research, Division of Population Health and Immunity, Melbourne, VIC, 3052, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Daniel P Poole
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, 3052, Australia.,Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Inoni Betuela
- Papua New Guinea Institute of Medical Research, Goroka and Madang, Papua New Guinea
| | - Danika L Hill
- Walter and Eliza Hall Institute of Medical Research, Division of Population Health and Immunity, Melbourne, VIC, 3052, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Peter M Siba
- Papua New Guinea Institute of Medical Research, Goroka and Madang, Papua New Guinea.,School of Veterinary and Biomedical Sciences, James Cook University, Townsville, QLD, 4811, Australia
| | - Diana S Hansen
- Walter and Eliza Hall Institute of Medical Research, Division of Population Health and Immunity, Melbourne, VIC, 3052, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Ivo Mueller
- Walter and Eliza Hall Institute of Medical Research, Division of Population Health and Immunity, Melbourne, VIC, 3052, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Emily M Eriksson
- Walter and Eliza Hall Institute of Medical Research, Division of Population Health and Immunity, Melbourne, VIC, 3052, Australia. .,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, 3052, Australia. .,The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Melbourne, VIC, 3052, Australia.
| |
Collapse
|
38
|
Naghavi-Alhosseini M, Tehrani M, Ajami A, Rafiei A, Taghvaei T, Vahedi-Larijani L, Hossein-Nataj H, Asgarian-Omran H. Tim-3 Up-regulation in Patients with Gastric Cancer and Peptic Ulcer Disease. Asian Pac J Cancer Prev 2017; 18:765-770. [PMID: 28441784 PMCID: PMC5464497 DOI: 10.22034/apjcp.2017.18.3.765] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background: T-cell immunoglobulin and mucin domain protein-3 (Tim-3), an inhibitory immunoregulatory receptor, has been recently implicated in tumor biology and tumor-associated immune suppression. In the present study, expression of Tim-3 was evaluated in gastric cancer (GC) and peptic ulcer disease (PUD) at both mRNA and protein levels. Methods: A total of 133 gastric tissue biopsies, comprising 43 from GC cases, 48 from PUD and 42 from non-ulcer dyspepsia (NUD) serving as controls were collected. Additionally, non-neoplastic adjacent tissue biopsies were also obtained from 6 patients with GC. Infection with Helicobacter pylori was determined by the rapid urease test for all participants and H&E staining was conducted for GC and PUD patients. Tim-3 relative mRNA expression was determined by SYBR Green based Real-Time PCR using β-actin as a reference gene. Tim-3 protein expression was also studied by immunohistochemistry in 7 GC, 7 PUD and 10 NUD tissue samples. Results: Tim-3 was expressed at higher levels in GC (p=0.030) and PUD (p=0.022) cases compared to he NUD group. Among paired samples obtained from gastric cancer patients, tumor tissues showed elevated Tim-3 expression (p=0.019) in comparison with adjacent non-neoplastic biopsies. Tim-3 mRNA findings were supported by detection of more Tim-3 protein in cancerous (p=0.002) and ulcerative (p=0.01) tissues than in controls. Tim-3 was similarly expressed in H. pylori positive and negative cases. Conclusion: Higher Tim-3 expression in patients with gastric cancer and peptic ulcer implies that it might be involved in immune regulation and establishment of these gastrointestinal diseases. Targeted immunotherapy by blocking of inhibitory receptors like Tim-3 could be a promising approach for gastric cancer treatment.
Collapse
Affiliation(s)
- Mahdieh Naghavi-Alhosseini
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Molecular and Cell Biology Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Monoclonal antibody binding to the macrophage-specific receptor sialoadhesin alters the phagocytic properties of human and mouse macrophages. Cell Immunol 2016; 312:51-60. [PMID: 27993350 DOI: 10.1016/j.cellimm.2016.11.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 11/15/2016] [Accepted: 11/28/2016] [Indexed: 12/17/2022]
Abstract
Sialoadhesin (Sn) is a surface receptor expressed on macrophages in steady state conditions, but during inflammation, Sn can be upregulated both on macrophages and on circulating monocytes. It was shown for different species that Sn becomes internalized after binding with monoclonal antibodies. These features suggest that Sn is a potential target for immunotherapies. In this study, human and mouse macrophages were treated with anti-Sn monoclonal antibodies or F(ab')2 fragments and the effect of their binding to Sn on phagocytosis was analyzed. Binding of antibodies to Sn resulted in delayed and reduced phagocytosis of fluorescent beads. No effect was observed on Fc-mediated phagocytosis or phagocytosis of bacteria by human macrophages. In contrast, an enhanced phagocytosis of bacteria by mouse macrophages was detected. These results showed that stimulation of Sn could have different effects on macrophage phagocytosis, depending both on the type of phagocytosis and cellular background.
Collapse
|
40
|
Homayouni V, Ganjalikhani-hakemi M, Rezaei A, Khanahmad H, Behdani M, Lomedasht FK. Preparation and characterization of a novel nanobody against T-cell immunoglobulin and mucin-3 (TIM-3). IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2016; 19:1201-1208. [PMID: 27917276 PMCID: PMC5126221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
OBJECTIVES As T-cell immunoglobulin and mucin domain 3 (TIM-3) is an immune regulatory molecule; its blocking or stimulating could alter the pattern of immune response towards a desired condition. Based on the unique features of nanobodies, we aimed to construct an anti-TIM-3 nanobody as an appropriate tool for manipulating immune responses for future therapeutic purposes. MATERIALS AND METHODS We immunized a camel with TIM-3 antigen and then, synthesized a VHH phage: mid library from its B cell's transcriptome using nested PCR. Library selection against TIM-3antigen was performed in three rounds of panning. Using phage-ELISA, the most reactive colonies were selected for sub-cloning in soluble protein expression vectors. The Nanobody was purified and confirmed with a nickel-nitrilotriacetic acid (Ni-NTA) column, SDS-PAGE and Western blotting. A flowcytometric analysis was performed to analyze the binding and biologic activities of theTIM-3 specific nanobody with TIM-3 expressing HL-60 and HEK cell lines. RESULTS Specific 15kD band representing for nanobody was observed on the gel and confirmed with Western blotting. The nanobody showed significant specific immune-reactivity against TIM-3 with a relatively high binding affinity. The nanobody significantly suppressed the proliferation of TIM-3 expressing HL-60 cell line. CONCLUSION Finally, we successfully prepared a functional anti-humanTIM-3 specific nanobody with a high affinity and an anti-proliferative activity on an AML cell line in vitro.
Collapse
Affiliation(s)
- Vida Homayouni
- Immunology Department, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mazdak Ganjalikhani-hakemi
- Immunology Department, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran,Corresponding author: Mazdak Ganjalikhani-hakemi. Immunology Department, Faculty of Medicine, Isfahan University of Medical Sciences. Isfahan, Iran. Tel: +98-31-37929082; Fax: +98-31-37929031;
| | - Abbas Rezaei
- Immunology Department, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Khanahmad
- Genetic Department, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahdi Behdani
- Biotechnology Research Center, Biotechnology Department, Venom & Bio-therapeutics Molecules Lab, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Kazemi Lomedasht
- Biotechnology Research Center, Biotechnology Department, Venom & Bio-therapeutics Molecules Lab, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
41
|
Kishimoto W, Nishikori M, Arima H, Miyoshi H, Sasaki Y, Kitawaki T, Shirakawa K, Kato T, Imaizumi Y, Ishikawa T, Ohno H, Haga H, Ohshima K, Takaori-Kondo A. Expression of Tim-1 in primary CNS lymphoma. Cancer Med 2016; 5:3235-3245. [PMID: 27709813 PMCID: PMC5119979 DOI: 10.1002/cam4.930] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/07/2016] [Accepted: 09/07/2016] [Indexed: 11/16/2022] Open
Abstract
Primary central nervous system lymphoma (PCNSL) is a distinct subtype of extranodal lymphoma with aggressive clinical course and poor outcome. As increased IL‐10/IL‐6 ratio is recognized in the cerebrospinal fluid (CSF) of PCNSL patients, we hypothesized that PCNSL might originate from a population of B cells with high IL‐10‐producing capacity, an equivalent of “regulatory B cells” in mice. We intended in this study to clarify whether Tim‐1, a molecule known as a marker for regulatory B cells in mice, is expressed in PCNSL. By immunohistochemical analysis, Tim‐1 was shown to be positive in as high as 54.2% of PCNSL (26 of 58 samples), while it was positive in 19.1% of systemic diffuse large B‐cell lymphoma (DLBCL) samples (17 of 89 samples; P < 0.001). Tim‐1 expression positively correlated with IL‐10 expression in PCNSL (Cramer's V = 0.55, P < 0.001), and forced expression of Tim‐1 in a PCNSL cell line resulted in increased IL‐10 secretion, suggesting that Tim‐1 is functionally linked with IL‐10 production in PCNSL. Moreover, soluble Tim‐1 was detectable in the CSF of PCNSL patients, and was suggested to parallel disease activity. In summary, PCNSL is characterized by frequent Tim‐1 expression, and its soluble form in CSF may become a useful biomarker for PCNSL.
Collapse
Affiliation(s)
- Wataru Kishimoto
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Momoko Nishikori
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Arima
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroaki Miyoshi
- Department of Pathology, Kurume University School of Medicine, Asahimachi, Kurume, Fukuoka, Japan
| | - Yuya Sasaki
- Department of Pathology, Kurume University School of Medicine, Asahimachi, Kurume, Fukuoka, Japan
| | - Toshio Kitawaki
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kotaro Shirakawa
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeharu Kato
- Department of Hematology, Nagasaki University Hospital, Sakamoto, Nagasaki, Japan
| | - Yoshitaka Imaizumi
- Department of Hematology, Nagasaki University Hospital, Sakamoto, Nagasaki, Japan
| | - Takayuki Ishikawa
- Department of Hematology, Kobe City Medical Center General Hospital, Kobe, Hyogo, Japan
| | - Hitoshi Ohno
- Department of Hematology, Tenri Hospital, Mishima-cho, Tenri, Nara, Japan
| | - Hironori Haga
- Department of Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Koichi Ohshima
- Department of Pathology, Kurume University School of Medicine, Asahimachi, Kurume, Fukuoka, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
42
|
Hiraishi Y, Nambu A, Shibui A, Nakanishi W, Yamaguchi S, Morita H, Iikura M, McKenzie AN, Matsumoto K, Sudo K, Yamasoba T, Nagase T, Nakae S. TIM-3 is not essential for development of airway inflammation induced by house dust mite antigens. Allergol Int 2016; 65:459-465. [PMID: 27209052 PMCID: PMC5074363 DOI: 10.1016/j.alit.2016.04.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/29/2016] [Accepted: 04/13/2016] [Indexed: 12/11/2022] Open
Abstract
Background T cell immunoglobulin domain and mucin domain-containing molecule 3 (TIM-3), which is preferentially expressed on Th1 cells rather than Th2 cells, is considered to be a negative regulator of Th1 cell function. This suggests that TIM-3 indirectly enhances Th2-type immune responses by suppressing Th1 cell function. Methods To investigate TIM-3's possible involvement in Th2-type acute and chronic airway inflammation, wild-type and TIM-3-deficient (TIM-3−/−) mice were sensitized and challenged with a house dust mite (HDM) extract. Airway inflammation and the number of inflammatory cells in bronchoalveolar lavage fluids (BALFs) in the mice were determined by histological analysis and with a hemocytometer, respectively. Expression of mRNA in the lungs was determined by quantitative PCR, while the levels of cytokines in the BALFs and IgE in sera were determined by ELISA. Results Despite constitutive expression of TIM-3 mRNA in the lungs, the number of eosinophils in bronchoalveolar lavage fluids (BALFs) and the score of pulmonary inflammation were comparable between wild-type and TIM-3−/− mice during both acute and chronic HDM-induced airway inflammation. On the other hand, the number of lymphocytes in the BALFs of TIM-3−/− mice was significantly increased compared with wild-type mice during HDM-induced chronic, but not acute, airway inflammation, while the levels of Th2 cytokines in the BALFs and HDM-specific IgG1 and IgG2a and total IgE in the sera were comparable in both groups. Conclusions Our findings indicate that, in mice, TIM-3 is not essential for development of HDM-induced acute or chronic allergic airway inflammation, although it appears to be involved in reduced lymphocyte recruitment during HDM-induced chronic allergic airway inflammation.
Collapse
|
43
|
Ocaña-Guzman R, Torre-Bouscoulet L, Sada-Ovalle I. TIM-3 Regulates Distinct Functions in Macrophages. Front Immunol 2016; 7:229. [PMID: 27379093 PMCID: PMC4904032 DOI: 10.3389/fimmu.2016.00229] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 05/26/2016] [Indexed: 12/28/2022] Open
Abstract
The transmembrane protein TIM-3 is a type I protein expressed by sub-types of lymphoid cells, such as lymphocytes Th1, Th17, Tc1, NK, as well as in myeloid cells. Scientific evidence indicates that this molecule acts as a negative regulator of T lymphocyte activation and that its expression is modified in viral infections or autoimmune diseases. In addition to evidence from lymphoid cells, the function of TIM-3 has been investigated in myeloid cells, such as monocytes, macrophages, and dendritic cells (DC), where studies have demonstrated that it can regulate cytokine production, cell activation, and the capture of apoptotic bodies. Despite these advances, the function of TIM-3 in myeloid cells and the molecular mechanisms that this protein regulates are not yet fully understood. This review examines the most recent evidence concerning the function of TIM-3 when expressed in myeloid cells, primarily macrophages, and the potential impact of that function on the field of basic immunology.
Collapse
Affiliation(s)
- Ranferi Ocaña-Guzman
- Laboratorio de Inmunología Integrativa, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, México City, México
| | - Luis Torre-Bouscoulet
- Departamento de Fisiología Respiratoria, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, México City, México
| | - Isabel Sada-Ovalle
- Laboratorio de Inmunología Integrativa, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, México City, México
| |
Collapse
|
44
|
Xu XG, Hu JF, Ma JX, Nie L, Shao T, Xiang LX, Shao JZ. Essential Roles of TIM-1 and TIM-4 Homologs in Adaptive Humoral Immunity in a Zebrafish Model. THE JOURNAL OF IMMUNOLOGY 2016; 196:1686-99. [PMID: 26792807 DOI: 10.4049/jimmunol.1501736] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 12/17/2015] [Indexed: 01/08/2023]
Abstract
TIM-1 and TIM-4 proteins have become increasingly attractive for their critical functions in immune modulation, particularly in CD4(+) Th2 cell activation. Thus, these proteins were hypothesized to regulate adaptive humoral immunity. However, further evidence is needed to validate this hypothesis. This study describes the molecular and functional characteristics of TIM-1 and TIM-4 homologs from a zebrafish (Danio rerio) model (D. rerio TIM [DrTIM]-1 and DrTIM-4). DrTIM-1 and DrTIM-4 were predominantly expressed in CD4(+) T cells and MHC class II(+) APCs under the induction of Ag stimulation. Blockade or knockdown of both DrTIM-1 and DrTIM-4 significantly decreased Ag-specific CD4(+) T cell activation, B cell proliferation, Ab production, and vaccinated immunoprotection against bacterial infection. This result suggests that DrTIM-1 and DrTIM-4 serve as costimulatory molecules required for the full activation of adaptive humoral immunity. DrTIM-1 was detected to be a trafficking protein located in the cytoplasm of CD4(+) T cells. It can translocate onto the cell surface under stimulation by TIM-4-expressing APCs, which might be a precise regulatory strategy for CD4(+) T cells to avoid self-activation before APCs stimulation. Furthermore, a unique alternatively spliced soluble DrTIM-4 variant was identified to exert a negative regulatory effect on the proliferation of CD4(+) T cells. The above findings highlight a novel costimulatory mechanism underlying adaptive immunity. This study enriches the current knowledge on TIM-mediated immunity and provides a cross-species understanding of the evolutionary history of costimulatory systems throughout vertebrate evolution.
Collapse
Affiliation(s)
- Xiao-Gang Xu
- College of Life Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China; Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou 310058, People's Republic of China; and
| | - Jing-Fang Hu
- College of Life Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China; Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou 310058, People's Republic of China; and
| | - Jun-Xia Ma
- College of Life Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China; Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou 310058, People's Republic of China; and
| | - Li Nie
- College of Life Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China; Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou 310058, People's Republic of China; and
| | - Tong Shao
- College of Life Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China; Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou 310058, People's Republic of China; and
| | - Li-Xin Xiang
- College of Life Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China; Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou 310058, People's Republic of China; and
| | - Jian-Zhong Shao
- College of Life Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China; Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou 310058, People's Republic of China; and Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, People's Republic of China
| |
Collapse
|
45
|
Awuah SG, Zheng YR, Bruno PM, Hemann MT, Lippard SJ. A Pt(IV) Pro-drug Preferentially Targets Indoleamine-2,3-dioxygenase, Providing Enhanced Ovarian Cancer Immuno-Chemotherapy. J Am Chem Soc 2015; 137:14854-7. [PMID: 26561720 PMCID: PMC4772771 DOI: 10.1021/jacs.5b10182] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Expression of indoleamine-2,3-dioxygenase (IDO), an immunosuppressive enzyme in human tumors, leads to immune evasion and tumor tolerance. IDO is therefore a tumor immunotherapeutic target, and several IDO inhibitors are currently undergoing clinical trials. IDO inhibitors can enhance the efficacy of common cancer chemotherapeutics. Here we investigate Pt(IV)-(D)-1-methyltryptophan conjugates 1 and 2 for combined immunomodulation and DNA cross-link-triggered apoptosis for cancer "immuno-chemotherapy". Compound 2 effectively kills hormone-dependent, cisplatin-resistant human ovarian cancer cells, inhibiting IDO by transcriptional deregulation of the autocrine-signaling loop IDO-AHR-IL6, which blocks kynurenine production and promotes T-cell proliferation. Additionally, 1 and 2 display low toxicity in mice and are stable in blood. To our knowledge, this construct is the first Pt drug candidate with immune checkpoint blockade properties.
Collapse
Affiliation(s)
- Samuel G Awuah
- Department of Chemistry, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| | - Yao-Rong Zheng
- Department of Chemistry, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| | - Peter M Bruno
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| | - Michael T Hemann
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| | - Stephen J Lippard
- Department of Chemistry, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| |
Collapse
|
46
|
Ebrahimi M, Kazemi T, Ganjalikhani-hakemi M, Majidi J, khanahmad H, Rahimmanesh I, Homayouni V, Kohpayeh S. Development of a Stable Cell Line, Overexpressing Human T-cell Immunoglobulin Mucin 1. IRANIAN JOURNAL OF BIOTECHNOLOGY 2015; 13:25-31. [PMID: 28959306 PMCID: PMC5492232 DOI: 10.15171/ijb.1350] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Revised: 11/12/2015] [Accepted: 11/24/2015] [Indexed: 02/05/2023]
Abstract
BACKGROUND Recent researches have demonstrated that human T-cell immunoglobulin mucin 1 (TIM-1) glycoprotein plays important roles in regulation of autoimmune and allergic diseases, as well as in tumor immunity and response to viral infections. Therefore, targeting TIM-1 could be a potential therapeutic approach against such diseases. OBJECTIVES In this study, we aimed to express TIM-1 protein on Human Embryonic kidney (HEK) 293T cell line in order to have an available source of the TIM-1 antigen. MATERIALS AND METHODS The cDNA was synthesized after RNA extraction from peripheral blood mononuclear cells (PBMC) and TIM-1 cDNA was amplified by PCR with specific primers. The PCR product was cloned in pcDNA™3.1/Hygro (+) and transformed in Escherichia coli TOP 10 F'. After cloning, authenticity of DNA sequence was checked and expressed in HEK 293T cells. Finally, expression of TIM-1 was analyzed by flow cytometry and real-time PCR. RESULTS The result of DNA sequencing demonstrated correctness of TIM-1 DNA sequence. The flow cytometry results indicated that TIM-1 was expressed in about 90% of transfected HEK 293T cells. The real-time PCR analysis showed TIM-1 mRNA expression increased 195-fold in transfected cells compared with un-transfected cells. CONCLUSIONS Findings of present study demonstrated the successful cloning and expression of TIM-1 on HEK 293T cells. These cells could be used as an immunogenic source for production of specific monoclonal antibodies, nanobodies and aptamers against human TIM-1.
Collapse
Affiliation(s)
- Mina Ebrahimi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Jafar Majidi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein khanahmad
- Department of Genetic and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ilnaz Rahimmanesh
- Department of Genetic and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Vida Homayouni
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shirin Kohpayeh
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
47
|
Echbarthi M, Zonca M, Mellwig R, Schwab Y, Kaplan G, DeKruyff RH, Roda-Navarro P, Casasnovas JM. Distinct Trafficking of Cell Surface and Endosomal TIM-1 to the Immune Synapse. Traffic 2015; 16:1193-207. [PMID: 26332704 DOI: 10.1111/tra.12329] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 08/26/2015] [Accepted: 08/26/2015] [Indexed: 01/31/2023]
Abstract
The T cell costimulatory molecule TIM-1 (T cell/transmembrane, mucin and immunoglobulin domain protein 1) sorts mainly to endosomes in lymphoid cells. At difference from the cell surface protein, endosomal TIM-1 translocates to the immune synapse (IS), where it can contribute to antigen-dependent T cell costimulation. TIM-1 ligands increase the amount of cell surface protein, preventing its traffic to the IS. The bipolar sorting of TIM-1 observed during IS formation is determined by differences in its subcellular location, and probably modulates antigen-driven immune responses.
Collapse
Affiliation(s)
- Meriem Echbarthi
- Macromolecule Structure, Centro Nacional de Biotecnología, CNB-CSIC, Madrid, 28049, Spain.,Current address: Department of Chemistry and Molecular Biology, University of Gothenburg, Göteborg, Sweden
| | - Manuela Zonca
- Macromolecule Structure, Centro Nacional de Biotecnología, CNB-CSIC, Madrid, 28049, Spain
| | | | | | - Gerardo Kaplan
- Food and Drug Administration, Center for Biologics Evaluation and Research, Silver Spring, MD, 20993, USA
| | | | - Pedro Roda-Navarro
- Department of Immunology, School of Medicine, Complutense University, 12 de Octubre Health Research Institute, Madrid, Spain
| | - Jose M Casasnovas
- Macromolecule Structure, Centro Nacional de Biotecnología, CNB-CSIC, Madrid, 28049, Spain
| |
Collapse
|
48
|
Abstract
Tim-3 is a member of the T cell immunoglobulin and mucin domain (Tim) family of proteins, which are expressed by several cell types in the immune system, including CD4 and CD8 T cells activated under certain conditions. These molecules are generally thought to act as receptors for multiple ligands and thus to function by engaging intracellular signaling pathways in a ligand-dependent manner. In recent years, the function of the Tim-3 protein has been studied in some detail, particularly with respect to its role in the regulation of CD4 and CD8 T cell responses. Here, we review the structural features of Tim-3, known ligands for this molecule and the links established between Tim-3 and signal transduction pathways. In addition, we review the current literature regarding the role of Tim-3 in the regulation of effector responses by CD4 and CD8 T cells. Overall, findings published thus far strongly support the conclusion that Tim-3 functions to inhibit T cell responses, particularly under conditions involving chronic stimulation. Conversely, some reports have provided evidence that Tim-3 can stimulate T cells under conditions involving acute stimulation, suggesting that the role of Tim-3 may vary depending on context. Further study of Tim-3 is likely to advance our understanding of how CD4 and CD8 T cell responses are regulated and could uncover novel approaches for manipulating T cell function for therapeutic benefit.
Collapse
|
49
|
Baghdadi M, Takeuchi S, Wada H, Seino KI. Blocking monoclonal antibodies of TIM proteins as orchestrators of anti-tumor immune response. MAbs 2014; 6:1124-32. [PMID: 25517298 DOI: 10.4161/mabs.32107] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Monoclonal antibody (mAb)-based treatment of cancer has a significant effect on current practice in medical oncology, and is considered now as one of the most successful therapeutic strategies for cancer treatment. MAbs are designed to initiate or enhance anti-tumor immune responses, which can be achieved by either blocking inhibitory immune checkpoint molecules or triggering activating receptors. TIM gene family members are type-I surface molecules expressed in immune cells, and play important roles in the regulation of both innate and adaptive arms of the immune system. Therapeutic strategies based on anti-TIMs mAbs have shown promising results in experimental tumor models, and synergistic combinations of anti-TIMs mAbs with cancer vaccines, adoptive T-cell therapy, radiotherapy and chemotherapy will have great impact on cancer treatment in future clinical development.
Collapse
Affiliation(s)
- Muhammad Baghdadi
- a Division of Immunobiology; Institute for Genetic Medicine ; Hokkaido University ; Sapporo , Japan
| | | | | | | |
Collapse
|
50
|
Ayala-Lujan JL, Vijayakumar V, Gong M, Smith R, Santiago AE, Ruiz-Perez F. Broad spectrum activity of a lectin-like bacterial serine protease family on human leukocytes. PLoS One 2014; 9:e107920. [PMID: 25251283 PMCID: PMC4176022 DOI: 10.1371/journal.pone.0107920] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 08/25/2014] [Indexed: 11/21/2022] Open
Abstract
The serine protease autotransporter from Enterobacteriaceae (SPATE) family, which number more than 25 proteases with apparent diverse functions, have been phylogenetically divided into two distinct classes, designated 1 and 2. We recently demonstrated that Pic and Tsh, two members of the class-2 SPATE family produced by intestinal and extraintestinal pathogenic E. coli, were able to cleave a number of O-glycosylated proteins on neutrophils and lymphocytes resulting in impaired leukocyte functions. Here we show that most members of the class-2 SPATE family have lectin-like properties and exhibit differential protease activity reliant on glycoprotein type and cell lineage. Protease activity was seen in virtually all tested O-glycosylated proteins including CD34, CD55, CD164, TIM1, TIM3, TIM4 and C1-INH. We also show that although SPATE proteins bound and cleaved glycoproteins more efficiently on granulocytes and monocytes, they also targeted glycoproteins on B, T and natural killer lymphocytes. Finally, we found that the characteristic domain-2 of class-2 SPATEs is not required for glycoprotease activity, but single amino acid mutations in Pic domain-1 to those residues naturally occurring in domain-1 of SepA, were sufficient to hamper Pic glycoprotease activity. This study shows that most class-2 SPATEs have redundant activities and suggest that they may function as immunomodulators at several levels of the immune system.
Collapse
Affiliation(s)
- Jorge Luis Ayala-Lujan
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- Unidad Academica de Ciencias Quimicas, Universidad Autonoma de Zacatecas, Zacatecas, Mexico
| | - Vidhya Vijayakumar
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- Department of Immunology and Microbiology, University of Maryland at Baltimore, Baltimore, Maryland, United States of America
| | - Mei Gong
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Rachel Smith
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Araceli E. Santiago
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Fernando Ruiz-Perez
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|