1
|
Naylon SH, Richaud AD, Zhao G, Bui L, Dufresne CP, Wu CJ, Wangpaichitr M, Savaraj N, Roche SP. A platform of ADAPTive scaffolds: development of CDR-H3 β-hairpin mimics into covalent inhibitors of the PD1/PDL1 immune checkpoint. RSC Chem Biol 2024; 5:d4cb00174e. [PMID: 39552936 PMCID: PMC11562385 DOI: 10.1039/d4cb00174e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/30/2024] [Indexed: 11/19/2024] Open
Abstract
Aberrant and dysregulated protein-protein interactions (PPIs) drive a significant number of human diseases, which is why they represent a major class of targets in drug discovery. Although a number of high-affinity antibody-based drugs have emerged in this therapeutic space, the discovery of smaller PPI inhibitors is lagging far behind, underscoring the need for novel scaffold modalities. To bridge this gap, we introduce a biomimetic platform technology - adaptive design of antibody paratopes into therapeutics (ADAPT) - that enables the paratope-forming binding loops of antibodies to be crafted into large β-hairpin scaffolds (ADAPTins). In this study, we describe a novel strategy for engineering native CDR-H3 "hot loops" with varying sequences, lengths, and rigidity into ADAPTins, ultimately transforming these compounds into irreversible covalent inhibitors. A proof-of-concept was established by creating a series of ADAPTin blockers of the PD1:PDL1 immune checkpoint PPI (blocking activity EC50 < 0.3 μM) which were subsequently modified into potent covalent PD1 inhibitors. The compelling rate of stable and folded ADAPTins above physiological temperature (21 out of 29) obtained across six different scaffolds suggests that the platform technology could provide a novel opportunity for high-quality peptide display and biological screening.
Collapse
Affiliation(s)
- Sarah H Naylon
- Department of Chemistry and Biochemistry, Florida Atlantic University Boca Raton Florida 33431 USA
| | - Alexis D Richaud
- Department of Chemistry and Biochemistry, Florida Atlantic University Boca Raton Florida 33431 USA
| | - Guangkuan Zhao
- Department of Chemistry and Biochemistry, Florida Atlantic University Boca Raton Florida 33431 USA
| | - Linda Bui
- Department of Chemistry and Biochemistry, Florida Atlantic University Boca Raton Florida 33431 USA
| | | | - Chunjing J Wu
- University of Miami, Miller School of Medicine Miami Florida 33136 USA
| | | | - Niramol Savaraj
- University of Miami, Miller School of Medicine Miami Florida 33136 USA
| | - Stéphane P Roche
- Department of Chemistry and Biochemistry, Florida Atlantic University Boca Raton Florida 33431 USA
| |
Collapse
|
2
|
Bakhtiyaridovvombaygi M, Yazdanparast S, Kheyrandish S, Safdari SM, Amiri Samani F, Sohani M, Jaafarian AS, Damirchiloo F, Izadpanah A, Parkhideh S, Mikanik F, Roshandel E, Hajifathali A, Gharehbaghian A. Harnessing natural killer cells for refractory/relapsed non-Hodgkin lymphoma: biological roles, clinical trials, and future prospective. Biomark Res 2024; 12:66. [PMID: 39020411 PMCID: PMC11253502 DOI: 10.1186/s40364-024-00610-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/28/2024] [Indexed: 07/19/2024] Open
Abstract
Non-Hodgkin lymphomas (NHLs) are heterogeneous and are among the most common hematological malignancies worldwide. Despite the advances in the treatment of patients with NHLs, relapse or resistance to treatment is anticipated in several patients. Therefore, novel therapeutic approaches are needed. Recently, natural killer (NK) cell-based immunotherapy alone or in combination with monoclonal antibodies, chimeric antigen receptors, or bispecific killer engagers have been applied in many investigations for NHL treatment. The functional defects of NK cells and the ability of cancerous cells to escape NK cell-mediated cytotoxicity within the tumor microenvironment of NHLs, as well as the beneficial results from previous studies in the context of NK cell-based immunotherapy in NHLs, direct our attention to this therapeutic strategy. This review aims to summarize clinical studies focusing on the applications of NK cells in the immunotherapy of patients with NHL.
Collapse
Affiliation(s)
- Mehdi Bakhtiyaridovvombaygi
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somayeh Yazdanparast
- Department of Hematology and Blood Banking, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Setare Kheyrandish
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mehrab Safdari
- Departments of Hematology and Blood Transfusion, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fateme Amiri Samani
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran
| | - Mahsa Sohani
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Akram Sadat Jaafarian
- Departments of Hematology and Blood Transfusion, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fateme Damirchiloo
- Departments of Hematology and Blood Transfusion, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Izadpanah
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Parkhideh
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mikanik
- Laboratory Hematology and Blood Bank Department, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elham Roshandel
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abbas Hajifathali
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ahmad Gharehbaghian
- Laboratory Hematology and Blood Bank Department, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Pediatric Congenital Hematologic Disorders Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Nwozichi C, Ogunmuyiwa AO, Ojewale MO. Nurses' roles in CAR-T therapy for B-cell malignancies and managing associated cytokine release syndrome. Asia Pac J Oncol Nurs 2024; 11:100367. [PMID: 38304228 PMCID: PMC10831260 DOI: 10.1016/j.apjon.2023.100367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 12/23/2023] [Indexed: 02/03/2024] Open
Abstract
Introduction In recent times, significant innovations have been made in cancer immunotherapy. These innovations have yielded positive outcomes, including a substantial improvement in the clinical outcomes of cancer patients, especially in the B-cell setting involving patients with B-cell malignancies. Method This paper explores oncology nurses' actual and expanded roles in utilizing chimeric antigen receptor T-cell (CAR-T) therapy. Result CAR-T therapy is an exciting innovation in cancer treatment. However, this therapy is often associated with some mild to life-threatening side effects and toxicities, including cytokine release syndrome (CRS). Unfortunately, nurses lack adequate standardized guidelines for monitoring and managing patients with CRS. This paper explains oncology nurses' actual and expanded roles in utilizing CAR-T therapy in treating B-cell malignancies based on experience and published data. Discussion Nurses' responsibilities for patients experiencing CAR-T toxicities with a particular focus on CRS during treatment are discussed.
Collapse
|
4
|
Zajac KK, Malla S, Babu RJ, Raman D, Tiwari AK. Ethnic disparities in the immune microenvironment of triple negative breast cancer and its role in therapeutic outcomes. Cancer Rep (Hoboken) 2023; 6 Suppl 1:e1779. [PMID: 36632988 PMCID: PMC10440847 DOI: 10.1002/cnr2.1779] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 01/13/2023] Open
Abstract
In 2020, newly diagnosed breast cancer (BC) cases surpassed that of lung cancer among women, making it the most common female cancer globally. In spite of recent increases in incidence rates, mortality due to BC has declined since 1989. These declines have been attributed to advancements in treatment modalities as well as increased mammography surveillance. Despite these advances, African American (AA) women are 40% more likely to die from BC than Caucasian women. Multifactorial etiology has been implicated in the disparity of BC mortality rates among AA women. As an example, AA women have a disproportionate incidence of triple negative breast cancer (TNBC), which has a poor prognosis and marginal treatment options. Increasingly, the tumor microenvironment (TME) has gained relevance as it relates to primary tumor progression, metastasis and treatment possibilities. The treatment outcomes or pathological complete response (pCR) in TNBC among AA women are affected by differences in TME. The TME of AA women exhibit several variances in acellular and cellular components associated with pro-tumorigenic effects. For example, increased levels of the adipocyte-related hormone, resistin, the pro-inflammatory cytokine, IL-6, and the CC chemokine, CCL2, within the TME of AA women gives rise to an increased density of M2 macrophages, also known as tumor-associated macrophages. Elevated levels of vascular endothelial growth factor in the TME of AA women increase the vascular density or vascularity, which facilitate aggressive tumor growth and metastasis. Furthermore, a pro-tumorigenic TME is supported by increased levels of the CXC chemokine, CXCL12 that results in the recruitment of regulatory T lymphocytes (Tregs ). Due to these and other differences in the TME of AA women, precision oncology can target specific aspects of the TME that may contribute to a poorer prognosis. In addition to the discrepancies in the TME, AA women face socio-economic barriers that limit their ability to access state-of-the-art, novel therapies against metastatic TNBC. In this review, we will provide a brief overview of the tumor immune microenvironment, immune-based treatment options for TNBC and their potential to decrease health disparities due to ethnicity.
Collapse
Affiliation(s)
- Kelsee K. Zajac
- Department of Pharmacology and Experimental TherapeuticsThe University of ToledoToledoOhioUSA
| | - Saloni Malla
- Department of Pharmacology and Experimental TherapeuticsThe University of ToledoToledoOhioUSA
| | - Ramapuram Jayachandra Babu
- Department of Drug Discovery and Development, Harrison School of PharmacyAuburn UniversityAuburnAlabamaUSA
| | - Dayanidhi Raman
- Department of Cell and Cancer BiologyUniversity of Toledo Health Science CampusToledoOhioUSA
| | - Amit K. Tiwari
- Department of Pharmacology and Experimental TherapeuticsThe University of ToledoToledoOhioUSA
- Department of Cell and Cancer BiologyUniversity of Toledo Health Science CampusToledoOhioUSA
| |
Collapse
|
5
|
Chatterjee D, Al Rimon R, Chowdhury UF, Islam MR. A multi-epitope based vaccine against the surface proteins expressed in cyst and trophozoite stages of parasite Entamoeba histolytica. J Immunol Methods 2023; 517:113475. [PMID: 37088358 DOI: 10.1016/j.jim.2023.113475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/12/2023] [Accepted: 04/19/2023] [Indexed: 04/25/2023]
Abstract
Entamoeba histolytica, an anaerobic parasite, infects humans and other primates and causes fatal diseases, such as amebiasis, amebic liver abscesses, and many others. Thousands of people are infected and dying due to the need for a proper protective cure, especially in poor sanitizing regions, such as Latin America, Asia, and Africa. Around 10% of the world population is infected by E. histolytica every year. Consequently, novel preventive approaches are required to eliminate the threats of the parasite. A designed vaccine targeting the exposed proteins that are common between cyst and trophozoite stages of the parasite's life cycle would be an effective way to repress the impact of the parasite. Therefore, an in silico bioinformatics approach was performed to design an effective vaccine targeting surface proteins common between both stages of the parasite's life cycle using B-cell and T-cell epitopes. The epitopes derived from the conserved portions of the proteins and their corresponding isomers specific to the parasite suggested that the vaccine could benefit cross-protection. Furthermore, the three-dimensional structure of the designed vaccine was modelled, refined, and validated using multiple bioinformatics tools. The physiological properties and solubility were also predicted using different algorithmic tools and found to be highly soluble in nature. The vaccine was found interactcted with TLR immune receptors, and the stability was observed via dynamics simulation. Codon optimization and cloning were performed for expression analysis. Immune simulation prediction anticipated significant immune responses with a high IgG and IgM antibodies expression, Th and Tc cells population, B-cell population, memory cells, INF-γ, and IL-2 cytokines. Therefore, the constructed multi-epitope putative vaccine can effectively neutralize the parasite's harmful effects.
Collapse
Affiliation(s)
- Dipankor Chatterjee
- Department of Biochemistry and Molecular Biology, University of Dhaka, Bangladesh
| | - Razoan Al Rimon
- Department of Biochemistry and Molecular Biology, University of Dhaka, Bangladesh
| | - Umar Faruq Chowdhury
- Department of Biochemistry and Molecular Biology, University of Dhaka, Bangladesh
| | | |
Collapse
|
6
|
Exosomes for Regulation of Immune Responses and Immunotherapy. JOURNAL OF NANOTHERANOSTICS 2022. [DOI: 10.3390/jnt3010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Exosomes are membrane-enveloped nanosized (30–150 nm) extracellular vesicles of endosomal origin produced by almost all cell types and encompass a multitude of functioning biomolecules. Exosomes have been considered crucial players of cell-to-cell communication in physiological and pathological conditions. Accumulating evidence suggests that exosomes can modulate the immune system by delivering a plethora of signals that can either stimulate or suppress immune responses, which have potential applications as immunotherapies for cancer and autoimmune diseases. Here, we discuss the current knowledge about the active biomolecular components of exosomes that contribute to exosomal function in modulating different immune cells and also how these immune cell-derived exosomes play critical roles in immune responses. We further discuss the translational potential of engineered exosomes as immunotherapeutic agents with their advantages over conventional nanocarriers for drug delivery and ongoing clinical trials.
Collapse
|
7
|
Shkair L, Garanina EE, Martynova EV, Kolesnikova AI, Arkhipova SS, Titova AA, Rizvanov AA, Khaiboullina SF. Immunogenic Properties of MVs Containing Structural Hantaviral Proteins: An Original Study. Pharmaceutics 2022; 14:pharmaceutics14010093. [PMID: 35056989 PMCID: PMC8779827 DOI: 10.3390/pharmaceutics14010093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 02/01/2023] Open
Abstract
Hemorrhagic fever with renal syndrome (HFRS) is an emerging infectious disease that remains a global public health threat. The highest incidence rate is among zoonotic disease cases in Russia. Most cases of HFRS are reported in the Volga region of Russia, which commonly identifies the Puumala virus (PUUV) as a pathogen. HFRS management is especially challenging due to the lack of specific treatments and vaccines. This study aims to develop new approaches for HFRS prevention. Our goal is to test the efficacy of microvesicles (MVs) as PUUV nucleocapsid (N) and glycoproteins (Gn/Gc) delivery vehicles. Our findings show that MVs could deliver the PUUV N and Gn/Gc proteins in vitro. We have also demonstrated that MVs loaded with PUUV proteins could elicit a specific humoral and cellular immune response in vivo. These data suggest that an MV-based vaccine could control HFRS.
Collapse
|
8
|
Ghosh C, Luong G, Sun Y. A snapshot of the PD-1/PD-L1 pathway. J Cancer 2021; 12:2735-2746. [PMID: 33854633 PMCID: PMC8040720 DOI: 10.7150/jca.57334] [Citation(s) in RCA: 162] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 01/23/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer cells can evade the attack from host immune systems via hijacking the regulatory circuits mediated by immune checkpoints. Therefore, reactivating the antitumor immunity by blockade of immune checkpoints is considered as a promising strategy to treat cancer. Programmed death protein 1 (PD-1) and its ligand programmed death-ligand 1 (PD-L1) are critical immune checkpoint proteins that responsible for negative regulation of the stability and the integrity of T-cell immune function. Anti-PD-1/PD-L1 drugs have been developed for immune checkpoint blockade and can induce clinical responses across different types of cancers, which provides a new hope to cure cancer. However, the patients' response rates to current anti-PD-1 or anti-PD-L1 therapies are still low and many initial responders finally develop resistance to these therapies. In this review, we provides a snapshot of the PD-1/PD-L1 molecular structure, mechanisms controlling their expression, signaling modulated by PD-1/PD-L1, current anti-PD-1/PD-L1 therapies, and the future perspectives to overcome the resistance.
Collapse
Affiliation(s)
- Chinmoy Ghosh
- Philips Institute for Oral Health Research, School of Dentistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Gary Luong
- Philips Institute for Oral Health Research, School of Dentistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Yue Sun
- Philips Institute for Oral Health Research, School of Dentistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
9
|
Banik S, Rakshit S, Sarkar K. The Role of STAT1 in T Helper Cell Differentiation during Breast Cancer Progression. J Breast Cancer 2021; 24:253-265. [PMID: 34190440 PMCID: PMC8250105 DOI: 10.4048/jbc.2021.24.e34] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 03/25/2021] [Accepted: 05/02/2021] [Indexed: 01/10/2023] Open
Abstract
Members of the signal transducer and activator of transcription (STAT) protein family are intracellular transcription factors that facilitate several facets of cellular immunity, proliferation, apoptosis, and differentiation. They are principally stimulated by membrane receptor-associated Janus kinases. Dysregulation of this pathway is often detected in primary tumors and hints at augmented angiogenesis, which enriches tumors persistence and immunosuppression. STAT proteins play indispensable roles in cytokine signaling and T helper (Th) cell differentiation. Among STAT proteins, STAT1 plays a vital role in interferon signaling, which initiates the expression of genes encoding proteins with antitumor and apoptotic roles. STAT1 signaling is essential for Th1 cell differentiation. Several studies have also shown the role of STAT1 as a tumor suppressor in breast cancer, which is the most common intrusive malignancy and the second most common cause of cancer death in women. Herein, we review the intricate STAT1-mediated molecular mechanisms associated with Th cell differentiation and anti-tumor function in breast cancer.
Collapse
Affiliation(s)
- Sayantan Banik
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, India
| | - Sudeshna Rakshit
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, India
| | - Koustav Sarkar
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, India.
| |
Collapse
|
10
|
Desmirean M, Rauch S, Jurj A, Pasca S, Iluta S, Teodorescu P, Berce C, Zimta AA, Turcas C, Tigu AB, Moldovan C, Paris I, Steinheber J, Richlitzki C, Constantinescu C, Sigurjonsson OE, Dima D, Petrushev B, Tomuleasa C. B Cells versus T Cells in the Tumor Microenvironment of Malignant Lymphomas. Are the Lymphocytes Playing the Roles of Muhammad Ali versus George Foreman in Zaire 1974? J Clin Med 2020; 9:jcm9113412. [PMID: 33114418 PMCID: PMC7693982 DOI: 10.3390/jcm9113412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/15/2020] [Accepted: 10/19/2020] [Indexed: 12/15/2022] Open
Abstract
Malignant lymphomas are a heterogeneous group of malignancies that develop both in nodal and extranodal sites. The different tissues involved and the highly variable clinicopathological characteristics are linked to the association between the lymphoid neoplastic cells and the tissues they infiltrate. The immune system has developed mechanisms to protect the normal tissue from malignant growth. In this review, we aim to explain how T lymphocyte-driven control is linked to tumor development and describe the tumor-suppressive components of the resistant framework. This manuscript brings forward a new insight with regard to intercellular and intracellular signaling, the immune microenvironment, the impact of therapy, and its predictive implications. A better understanding of the key components of the lymphoma environment is important to properly assess the role of both B and T lymphocytes, as well as their interplay, just as two legendary boxers face each other in a heavyweight title final, as was the case of Ali versus Foreman.
Collapse
Affiliation(s)
- Minodora Desmirean
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania; (M.D.); (S.R.); (A.J.); (S.P.); (S.I.); (P.T.); (C.T.); (J.S.); (C.R.); (C.C.)
- Department of Pathology, Constantin Papilian Military Hospital, 400124 Cluj Napoca, Romania;
| | - Sebastian Rauch
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania; (M.D.); (S.R.); (A.J.); (S.P.); (S.I.); (P.T.); (C.T.); (J.S.); (C.R.); (C.C.)
| | - Ancuta Jurj
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania; (M.D.); (S.R.); (A.J.); (S.P.); (S.I.); (P.T.); (C.T.); (J.S.); (C.R.); (C.C.)
| | - Sergiu Pasca
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania; (M.D.); (S.R.); (A.J.); (S.P.); (S.I.); (P.T.); (C.T.); (J.S.); (C.R.); (C.C.)
| | - Sabina Iluta
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania; (M.D.); (S.R.); (A.J.); (S.P.); (S.I.); (P.T.); (C.T.); (J.S.); (C.R.); (C.C.)
| | - Patric Teodorescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania; (M.D.); (S.R.); (A.J.); (S.P.); (S.I.); (P.T.); (C.T.); (J.S.); (C.R.); (C.C.)
| | - Cristian Berce
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania; (C.B.); (A.-A.Z.); (A.-B.T.); (C.M.); (B.P.)
| | - Alina-Andreea Zimta
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania; (C.B.); (A.-A.Z.); (A.-B.T.); (C.M.); (B.P.)
| | - Cristina Turcas
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania; (M.D.); (S.R.); (A.J.); (S.P.); (S.I.); (P.T.); (C.T.); (J.S.); (C.R.); (C.C.)
| | - Adrian-Bogdan Tigu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania; (C.B.); (A.-A.Z.); (A.-B.T.); (C.M.); (B.P.)
| | - Cristian Moldovan
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania; (C.B.); (A.-A.Z.); (A.-B.T.); (C.M.); (B.P.)
| | - Irene Paris
- Department of Pathology, Constantin Papilian Military Hospital, 400124 Cluj Napoca, Romania;
| | - Jakob Steinheber
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania; (M.D.); (S.R.); (A.J.); (S.P.); (S.I.); (P.T.); (C.T.); (J.S.); (C.R.); (C.C.)
| | - Cedric Richlitzki
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania; (M.D.); (S.R.); (A.J.); (S.P.); (S.I.); (P.T.); (C.T.); (J.S.); (C.R.); (C.C.)
| | - Catalin Constantinescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania; (M.D.); (S.R.); (A.J.); (S.P.); (S.I.); (P.T.); (C.T.); (J.S.); (C.R.); (C.C.)
- Department of Anesthesia and Intensive Care, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania
| | - Olafur Eysteinn Sigurjonsson
- The Blood Bank, Landspitali—The National University Hospital of Iceland, 101 Reykjavik, Iceland;
- School of Science and Engineering, Reykjavik University, 101 Reykjavik, Iceland
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, 400124 Cluj Napoca, Romania;
| | - Bobe Petrushev
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania; (C.B.); (A.-A.Z.); (A.-B.T.); (C.M.); (B.P.)
- Department of Pathology, Octavian Fodor Regional Institute of Gastroenterology and Hepatology, 400124 Cluj Napoca, Romania
| | - Ciprian Tomuleasa
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania; (M.D.); (S.R.); (A.J.); (S.P.); (S.I.); (P.T.); (C.T.); (J.S.); (C.R.); (C.C.)
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, 400124 Cluj Napoca, Romania;
- Correspondence: ; Tel.: +40741337489
| |
Collapse
|
11
|
Ding M, Baker D. Recent advances in high-throughput flow cytometry for drug discovery. Expert Opin Drug Discov 2020; 16:303-317. [PMID: 33054417 DOI: 10.1080/17460441.2021.1826433] [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: 02/07/2023]
Abstract
INTRODUCTION High-throughput flow cytometry (HTFC) has proven to be an important technology in drug discovery. The use of HTFC enables multi-parametric screening of suspension cells containing heterogenous cell populations and coated particles for screening proteins of interest. Novel targets, novel cell markers and compound clusters for drug development have been identified from HTFC screens. AREAS COVERED In this article, the authors focus on reviewing the recent HTFC applications reported during the last 5-6 years, including drug discovery screens and studies for immune, immune-oncology, infectious and inflammatory diseases. The main HTFC approaches, development of HTFC systems, and automated sample preparation systems for HTFC are also discussed. EXPERT OPINION The advance of HTFC technology coupled with automated sample acquisition and sample preparation has demonstrated its utility in screening large numbers of compounds using suspension cells, facilitated screening of disease-relevant human primary cells, and enabled deep understanding of mechanism of action by analyzing multiple parameters. The authors see HTFC as a very valuable tool in immune, immune-oncology, infectious and inflammatory diseases where immune cells play essential roles.
Collapse
Affiliation(s)
- Mei Ding
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - David Baker
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| |
Collapse
|
12
|
Davis JS, Ferreira D, Paige E, Gedye C, Boyle M. Infectious Complications of Biological and Small Molecule Targeted Immunomodulatory Therapies. Clin Microbiol Rev 2020; 33:e00035-19. [PMID: 32522746 PMCID: PMC7289788 DOI: 10.1128/cmr.00035-19] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The past 2 decades have seen a revolution in our approach to therapeutic immunosuppression. We have moved from relying on broadly active traditional medications, such as prednisolone or methotrexate, toward more specific agents that often target a single receptor, cytokine, or cell type, using monoclonal antibodies, fusion proteins, or targeted small molecules. This change has transformed the treatment of many conditions, including rheumatoid arthritis, cancers, asthma, and inflammatory bowel disease, but along with the benefits have come risks. Contrary to the hope that these more specific agents would have minimal and predictable infectious sequelae, infectious complications have emerged as a major stumbling block for many of these agents. Furthermore, the growing number and complexity of available biologic agents makes it difficult for clinicians to maintain current knowledge, and most review articles focus on a particular target disease or class of agent. In this article, we review the current state of knowledge about infectious complications of biologic and small molecule immunomodulatory agents, aiming to create a single resource relevant to a broad range of clinicians and researchers. For each of 19 classes of agent, we discuss the mechanism of action, the risk and types of infectious complications, and recommendations for prevention of infection.
Collapse
Affiliation(s)
- Joshua S Davis
- Department of Infectious Diseases and Immunology, John Hunter Hospital, Newcastle, NSW, Australia
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia
| | - David Ferreira
- School of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Emma Paige
- Department of Infectious Diseases, Alfred Hospital, Melbourne, VIC, Australia
| | - Craig Gedye
- School of Medicine, University of New South Wales, Sydney, NSW, Australia
- Department of Oncology, Calvary Mater Hospital, Newcastle, NSW, Australia
| | - Michael Boyle
- Department of Infectious Diseases and Immunology, John Hunter Hospital, Newcastle, NSW, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
13
|
Manda EC, Chirove F. Acute hepatitis B virus infection model within the host incorporating immune cells and cytokine responses. Theory Biosci 2019; 139:153-169. [PMID: 31650408 DOI: 10.1007/s12064-019-00305-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/01/2019] [Indexed: 02/06/2023]
Abstract
We formulate and analyze a within-host hepatitis B viral mathematical model for hepatitis B in the acute phase of infection. The model incorporates hepatocytes, hepatitis B virus, immune system cells and cytokine dynamics using a system of ordinary differential equations. We use the model to demonstrate the trends of the hepatitis B infection qualitatively without the effects of immune cells and cytokines. Using these trends, we tested the effects of incorporating the immune cells only and immune cells with cytokine responses at low and high inhibitions on the hepatitis B virus infection. Our results showed that it is impossible to have the immune cells work independently from cytokines when there is an acute hepatitis B virus infection. Therefore, our results suggest that incorporating immune cells and cytokine dynamics in the acute hepatitis B virus infection stage delays infection in the hepatocytes and excluding such dynamics speeds up infection during this phase. Results from this study are useful in developing strategies for control of hepatocellular carcinoma which is caused by hepatitis B virus infection.
Collapse
Affiliation(s)
| | - Faraimunashe Chirove
- University of KwaZulu-Natal, Pietermaritzburg, South Africa.,University of Johannesburg, Johannesburg, South Africa
| |
Collapse
|
14
|
Paaso A, Jaakola A, Syrjänen S, Louvanto K. From HPV Infection to Lesion Progression: The Role of HLA Alleles and Host Immunity. Acta Cytol 2019; 63:148-158. [PMID: 30783048 DOI: 10.1159/000494985] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 10/29/2018] [Indexed: 01/27/2023]
Abstract
Persistent high-risk human papillomavirus (HPV) infection has been associated with increased risk for cervical precancerous lesions and cancer. The host's genetic variability is known to play a role in the development of cervical cancer. The human leukocyte antigen (HLA) genes are highly polymorphic and have shown to be important risk determinants of HPV infection persistence and disease progression. HLA class I and II cell surface molecules regulate the host's immune system by presenting HPV-derived peptides to T-cells. The activation of T-cell response may vary depending on the HLA allele polymorphism. The engagement of the T-cell receptor with the HPV peptide-HLA complex to create an active costimulatory signal is essential for the activation of the T-cell response. Functional peptide presentation by both HLA class I and II molecules is needed to activate efficient helper and effector T-cell responses in HPV infection recognition and clearance. Some of these HLA risk alleles could also be used as preventive tools in the detection of HPV-induced cervical lesions and cancer. These HLA alleles, together with HPV vaccines, could potentially offer possible solutions for reducing HPV-induced cervical cancer as well as other HPV-related cancers.
Collapse
Affiliation(s)
- Anna Paaso
- Department of Oral Pathology, Institute of Dentistry, Faculty of Medicine, University of Turku, Turku, Finland,
- Department of Obstetrics and Gynecology, Turku University Hospital, University of Turku, Turku, Finland,
| | - Anna Jaakola
- Department of Obstetrics and Gynecology, Turku University Hospital, University of Turku, Turku, Finland
- Department of Obstetrics and Gynecology, Kymenlaakso Central Hospital, Kotka, Finland
| | - Stina Syrjänen
- Department of Oral Pathology, Institute of Dentistry, Faculty of Medicine, University of Turku, Turku, Finland
- Department of Pathology, Turku University Hospital, University of Turku, Turku, Finland
| | - Karolina Louvanto
- Department of Oral Pathology, Institute of Dentistry, Faculty of Medicine, University of Turku, Turku, Finland
- Department of Obstetrics and Gynecology, Turku University Hospital, University of Turku, Turku, Finland
| |
Collapse
|
15
|
A brief review of clinical trials involving manipulation of invariant NKT cells as a promising approach in future cancer therapies. Cent Eur J Immunol 2017; 42:181-195. [PMID: 28860937 PMCID: PMC5573892 DOI: 10.5114/ceji.2017.69361] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 12/20/2016] [Indexed: 12/14/2022] Open
Abstract
In the recent years researchers have put a lot of emphasis on the possible immunotherapeutic strategies able to target tumors. Many studies have proven that the key role in recognition and eradication of cancer cells, both for mice and humans, is being conducted by the invariant natural killer T-cells (NKT). This small subpopulation of lymphocytes can kill other cells, either directly or indirectly, through the natural killer cells’ (NK) activation. They can also swiftly release cytokines, causing the involvement of elements of the innate and acquired immune system. With the discovery of α-galactosylceramide (α-GalCer) – the first known agonist for iNKT cells – and its later subsequent analogs, it became possible to effectively stimulate iNKT cells, hence to keep control over the tumor progression. This article refers to the current knowledge concerning iNKT cells and the most important aspects of their antitumor activity. It also highlights the clinical trials that aim at increasing the amount of iNKT cells in general and in the microenvironment of the tumor. For sure, the iNKT-based immunotherapeutic approach holds a great potential and is highly probable to become a part of the cancer immunotherapy in the future.
Collapse
|
16
|
Yuan T, Jiang Y, Li M, Li W. Chronic hepatitis B surface antigen seroclearance-related immune factors. Hepatol Res 2017; 47:49-59. [PMID: 27084584 DOI: 10.1111/hepr.12726] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 04/01/2016] [Accepted: 04/12/2016] [Indexed: 12/23/2022]
Abstract
The ultimate aims of the treatment of hepatitis B virus infection are the loss of hepatitis B surface antigen (HBsAg) and hepatitis B surface antibody seroconversion. Unfortunately, these goals are rarely reached. Many factors are associated with HBsAg seroconversion, including genetic, immune, and viral factors. However, the mechanism of HBsAg seroclearance, and particularly the immune mechanism, is still difficult to elucidate. The immune factor interferon-α is currently the main antiviral therapy for chronic hepatitis B virus infection. However, a sustained shift from response of HBsAg to hepatitis B surface antibody seroconversion is rarely obtained. Recent studies have revealed that several of the newly identified immune factors are closely related to the removal of HBsAg. In this article, we review recent studies on these immune factors, their influence on hepatitis B progression, and HBsAg seroconversion.
Collapse
Affiliation(s)
- Ting Yuan
- Liver Diseases Research Center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yongfang Jiang
- Liver Diseases Research Center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mei Li
- Liver Diseases Research Center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Li
- Liver Diseases Research Center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
17
|
Ballweg IC, Frölich K, Fandrey E, Kliem H, Pfaffl MW. Comparison of the immune competence of Turopolje, German Landrace × Turopolje, and German Landrace × Pietrain pigs after PRRSV vaccination. Vet Immunol Immunopathol 2016; 174:35-44. [PMID: 27185261 DOI: 10.1016/j.vetimm.2016.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 03/27/2016] [Accepted: 04/04/2016] [Indexed: 11/16/2022]
Abstract
The competences of the immune systems of the ancient pig breed Turopolje (T×T), German Landrace × Turopolje (L×T) and 'modern' pig breed German Landrace × Pietrain (L×P) were compared in this study. All pigs were immunized with a modified live vaccine against 'Porcine Reproductive and Respiratory Syndrome' (PRRS) virus (Ingelvac PRRS MLV(®)) to simulate an infection. Antibody production against PRRS MLV was evaluated in serum. Elimination of the viral infectious fragments during the experimental period was monitored in serum, leukocytes and tonsils by RT-qPCR. Furthermore relevant immune marker genes were quantified either on gene expression level using RT-qPCR [toll like receptor (TLR) 7, TLR8, TRAF6, CD163, SIGLEC1, CD4, CD8, CD14, CD19, tumor necrosis factor alpha (TNFα), interleukin (IL) 1, IL2, IL6, IL12], and on protein level using ELISA [interleukin (IL)-1, IL-2, IL-6, and IL-12]. The three breeds showed individual inactivation efficiencies as a reaction to the PRRS MLV vaccination. T×T eliminated the virus in serum within 16 days, followed by L×T (28 days) and L×P (36 days). The antibody titers against PRRS MLV of L×T and L×P were significantly higher compared to T×T (p<0.05). The gene expression data and protein analysis of interleukins revealed that T×T reacted with a type 1 immune response. In contrast, the two other breeds (L×T and L×P) showed a type 2 immune response, which resulted in the higher synthesis of B-cells and an increased concentration of specific anti-PRRS MLV antibodies.
Collapse
Affiliation(s)
- I C Ballweg
- Physiology Weihenstephan, School of Life Sciences, Technische Universität München, 85354 Freising, Weihenstephan, Germany
| | - K Frölich
- Arche Warder, Zentrum für alte Haus-und Nutztierrassen e.V., 24646 Warder, Germany
| | - E Fandrey
- Manhagen-Fuhlenau GbR, 24631 Langwedel, Germany
| | - H Kliem
- Physiology Weihenstephan, School of Life Sciences, Technische Universität München, 85354 Freising, Weihenstephan, Germany.
| | - M W Pfaffl
- Physiology Weihenstephan, School of Life Sciences, Technische Universität München, 85354 Freising, Weihenstephan, Germany
| |
Collapse
|
18
|
Ito H, Ando T, Ando K, Ishikawa T, Saito K, Moriwaki H, Seishima M. Induction of hepatitis B virus surface antigen-specific cytotoxic T lymphocytes can be up-regulated by the inhibition of indoleamine 2, 3-dioxygenase activity. Immunology 2014; 142:614-23. [PMID: 24580128 DOI: 10.1111/imm.12274] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 02/19/2014] [Accepted: 02/24/2014] [Indexed: 12/12/2022] Open
Abstract
Cytotoxic T lymphocytes (CTLs) are thought to be major effectors involved in viral clearance during acute infections, including hepatitis B virus (HBV) infection. A persistent HBV infection is characterized by a lack of or a weak CTL response to HBV, which may be reflective of tolerance to HBV. Efficient induction of HBV-specific CTLs leads to the clearance of HBV in patients with a chronic HBV infection. Previously, we reported that α-galactosylceramide (α-GalCer), a specific natural killer T (NKT) cell agonist, enhanced the induction of HBV surface antigen (HBsAg)-specific CTLs. In the present study, we found that inhibition of indoleamine 2,3-dioxygenase (IDO) activity enhanced the induction of HBsAg-specific CTLs after immunization with HBsAg and α-GalCer. The administration of HBsAg and α-GalCer increased the production of interleukin-2 and interleukin-12b, which are crucial for the induction of HBsAg-specific CTLs. The production of these cytokines was more strongly enhanced in IDO knockout mice compared with wild-type mice. In addition, α-GalCer induced the production of IDO in CD11b(+) cells, and these cells inhibited proliferation of HBsAg-specific CTLs. Our results lead to strategies for improving the induction of HBsAg-specific CTLs.
Collapse
Affiliation(s)
- Hiroyasu Ito
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | | | | | | | | | | | | |
Collapse
|
19
|
Liu Y, Song M, Che TM, Lee JJ, Bravo D, Maddox CW, Pettigrew JE. Dietary plant extracts modulate gene expression profiles in ileal mucosa of weaned pigs after an Escherichia coli infection. J Anim Sci 2014; 92:2050-62. [PMID: 24663182 DOI: 10.2527/jas.2013-6422] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
This study was conducted to characterize the effects of infection with a pathogenic F-18 Escherichia coli and 3 different plant extracts on gene expression of ileal mucosa in weaned pigs. Weaned pigs (total = 64, 6.3 ± 0.2 kg BW, and 21-d old) were housed in individual pens for 15 d, 4 d before and 11 d after the first inoculation (d 0). Treatments were in a 2 × 4 factorial arrangement: with or without an F-18 E. coli challenge and 4 diets (a nursery basal, control diet [CON], 10 ppm of capsicum oleoresin [CAP], garlic botanical [GAR], or turmeric oleoresin [TUR]). Results reported elsewhere showed that the plant extracts reduced diarrhea in challenged pigs. Total RNA (4 pigs/treatment) was extracted from ileal mucosa of pigs at d 5 post inoculation. Double-stranded cDNA was amplified, labeled, and further hybridized to the microarray, and data were analyzed in R. Differential gene expression was tested by fitting a mixed linear model in a 2 × 4 factorial ANOVA. Bioinformatics analysis was conducted by DAVID Bioinformatics Resources 6.7 (DAVID; National Institute of Allergy and Infectious Diseases [NIAID, NIH], http://david.abcc.ncifcrf.gov). The E. coli infection altered (P < 0.05) the expression of 240 genes in pigs fed the CON (148 up- and 92 down-regulated). Compared with the infected CON, feeding CAP, GAR, or TUR altered (P < 0.05) the expression of 52 genes (18 up, 34 down), 117 genes (34 up- and 83 down-regulated), or 84 genes (16 up- and 68 down-regulated), respectively, often counteracting the effects of E. coli. The E. coli infection up-regulated (P < 0.05) the expression of genes related to the activation of immune response and complement and coagulation cascades, but down-regulated (P < 0.05) the expression of genes involved in protein synthesis and accumulation. Compared with the CON, feeding CAP and GAR increased (P < 0.05) the expression of genes related to integrity of membranes in infected pigs, indicating enhanced gut mucosa health. Moreover, feeding all 3 plant extracts reduced (P < 0.05) the expression of genes associated with antigen presentation or other biological processes of immune responses, indicating they attenuated overstimulation of immune responses caused by E. coli. These findings may explain why diarrhea was reduced and clinical immune responses were ameliorated in infected pigs fed plant extracts. In conclusion, plant extracts altered the expression of genes in ileal mucosa of E. coli-infected pigs, perhaps leading to the reduction in diarrhea reported previously.
Collapse
Affiliation(s)
- Y Liu
- Department of Animal Sciences and
| | | | | | | | | | | | | |
Collapse
|
20
|
Liu Y, Teige A, Mondoc E, Ibrahim S, Holmdahl R, Issazadeh-Navikas S. Endogenous collagen peptide activation of CD1d-restricted NKT cells ameliorates tissue-specific inflammation in mice. J Clin Invest 2010; 121:249-64. [PMID: 21157037 DOI: 10.1172/jci43964] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Accepted: 10/27/2010] [Indexed: 12/13/2022] Open
Abstract
NKT cells in the mouse recognize antigen in the context of the MHC class I-like molecule CD1d and play an important role in peripheral tolerance and protection against autoimmune and other diseases. NKT cells are usually activated by CD1d-presented lipid antigens. However, peptide recognition in the context of CD1 has also been documented, although no self-peptide ligands have been reported to date. Here, we have identified an endogenous peptide that is presented by CD1d to activate mouse NKT cells. This peptide, the immunodominant epitope from mouse collagen type II (mCII707-721), was not associated with either MHC class I or II. Activation of CD1d-restricted mCII707-721-specific NKT cells was induced via TCR signaling and classical costimulation. In addition, mCII707-721-specific NKT cells induced T cell death through Fas/FasL, in an IL-17A-independent fashion. Moreover, mCII707-721-specific NKT cells suppressed a range of in vivo inflammatory conditions, including delayed-type hypersensitivity, antigen-induced airway inflammation, collagen-induced arthritis, and EAE, which were all ameliorated by mCII707-721 vaccination. The findings presented here offer new insight into the intrinsic roles of NKT cells in health and disease. Given the results, endogenous collagen peptide activators of NKT cells may offer promise as novel therapeutics in tissue-specific autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Yawei Liu
- Neuroinflammation Unit, Biotech Research and Innovation Centre, University of Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|