101
|
Dertschnig S, Evans P, Santos E Sousa P, Manzo T, Ferrer IR, Stauss HJ, Bennett CL, Chakraverty R. Graft-versus-host disease reduces lymph node display of tissue-restricted self-antigens and promotes autoimmunity. J Clin Invest 2020; 130:1896-1911. [PMID: 31917684 PMCID: PMC7108931 DOI: 10.1172/jci133102] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 01/03/2020] [Indexed: 12/28/2022] Open
Abstract
Acute graft-versus-host disease (GVHD) is initially triggered by alloreactive T cells, which damage peripheral tissues and lymphoid organs. Subsequent transition to chronic GVHD involves the emergence of autoimmunity, although the underlying mechanisms driving this process are unclear. Here, we tested the hypothesis that acute GVHD blocks peripheral tolerance of autoreactive T cells by impairing lymph node (LN) display of peripheral tissue–restricted antigens (PTAs). At the initiation of GVHD, LN fibroblastic reticular cells (FRCs) rapidly reduced expression of genes regulated by DEAF1, an autoimmune regulator-like transcription factor required for intranodal expression of PTAs. Subsequently, GVHD led to the selective elimination of the FRC population, and blocked the repair pathways required for its regeneration. We used a transgenic mouse model to show that the loss of presentation of an intestinal PTA by FRCs during GVHD resulted in the activation of autoaggressive T cells and gut injury. Finally, we show that FRCs normally expressed a unique PTA gene signature that was highly enriched for genes expressed in the target organs affected by chronic GVHD. In conclusion, acute GVHD damages and prevents repair of the FRC network, thus disabling an essential platform for purging autoreactive T cells from the repertoire.
Collapse
Affiliation(s)
- Simone Dertschnig
- UCL Cancer Institute, and.,Institute of Immunity and Transplantation, London, United Kingdom
| | - Pamela Evans
- UCL Cancer Institute, and.,Institute of Immunity and Transplantation, London, United Kingdom
| | - Pedro Santos E Sousa
- UCL Cancer Institute, and.,Institute of Immunity and Transplantation, London, United Kingdom
| | | | - Ivana R Ferrer
- UCL Cancer Institute, and.,Institute of Immunity and Transplantation, London, United Kingdom
| | - Hans J Stauss
- Institute of Immunity and Transplantation, London, United Kingdom
| | - Clare L Bennett
- UCL Cancer Institute, and.,Institute of Immunity and Transplantation, London, United Kingdom
| | - Ronjon Chakraverty
- UCL Cancer Institute, and.,Institute of Immunity and Transplantation, London, United Kingdom
| |
Collapse
|
102
|
Weimershaus M, Mauvais FX, Evnouchidou I, Lawand M, Saveanu L, van Endert P. IRAP Endosomes Control Phagosomal Maturation in Dendritic Cells. Front Cell Dev Biol 2020; 8:585713. [PMID: 33425891 PMCID: PMC7793786 DOI: 10.3389/fcell.2020.585713] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/04/2020] [Indexed: 01/08/2023] Open
Abstract
Dendritic cells (DCs) contribute to the immune surveillance by sampling their environment through phagocytosis and endocytosis. We have previously reported that, rapidly following uptake of extracellular antigen into phagosomes or endosomes in DCs, a specialized population of storage endosomes marked by Rab14 and insulin-regulated aminopeptidase (IRAP) is recruited to the nascent antigen-containing compartment, thereby regulating its maturation and ultimately antigen cross-presentation to CD8+ T lymphocytes. Here, using IRAP–/– DCs, we explored how IRAP modulates phagosome maturation dynamics and cross-presentation. We find that in the absence of IRAP, phagosomes acquire more rapidly late endosomal markers, are more degradative, and show increased microbicidal activity. We also report evidence for a role of vesicle trafficking from the endoplasmic reticulum (ER)–Golgi intermediate compartment to endosomes for the formation or stability of the IRAP compartment. Moreover, we dissect the dual role of IRAP as a trimming peptidase and a critical constituent of endosome stability. Experiments using a protease-dead IRAP mutant and pharmacological IRAP inhibition suggest that IRAP expression but not proteolytic activity is required for the formation of storage endosomes and for DC-typical phagosome maturation, whereas proteolysis is required for fully efficient cross-presentation. These findings identify IRAP as a key factor in cross-presentation, trimming peptides to fit the major histocompatibility complex class-I binding site while preventing their destruction through premature phagosome maturation.
Collapse
Affiliation(s)
- Mirjana Weimershaus
- Institut National de la Santé et de la Recherche Médicale, Unité 1151, Université de Paris, Centre National de la Recherche Scientifique, UMR 8253, Paris, France
| | - François-Xavier Mauvais
- Institut National de la Santé et de la Recherche Médicale, Unité 1151, Université de Paris, Centre National de la Recherche Scientifique, UMR 8253, Paris, France
| | - Irini Evnouchidou
- Institut National de la Santé et de la Recherche Médicale, Unité 1151, Université de Paris, Centre National de la Recherche Scientifique, UMR 8253, Paris, France.,Inovarion, Paris, France
| | - Myriam Lawand
- Institut National de la Santé et de la Recherche Médicale, Unité 1151, Université de Paris, Centre National de la Recherche Scientifique, UMR 8253, Paris, France
| | - Loredana Saveanu
- Institut National de la Santé et de la Recherche Médicale, Unité 1151, Université de Paris, Centre National de la Recherche Scientifique, UMR 8253, Paris, France
| | - Peter van Endert
- Institut National de la Santé et de la Recherche Médicale, Unité 1151, Université de Paris, Centre National de la Recherche Scientifique, UMR 8253, Paris, France
| |
Collapse
|
103
|
Zhang Q, Huang W, Yuan M, Li W, Hua L, Yang Z, Gao F, Li S, Ye C, Chen Y, He J, Sun W, Yang X, Bai H, Ma Y. Employing ATP as a New Adjuvant Promotes the Induction of Robust Antitumor Cellular Immunity by a PLGA Nanoparticle Vaccine. ACS APPLIED MATERIALS & INTERFACES 2020; 12:54399-54414. [PMID: 33215918 DOI: 10.1021/acsami.0c15522] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Tumor vaccines based on synthetic human papillomavirus (HPV) oncoprotein E7 and/or E6 peptides have shown encouraging results in preclinical model studies and human clinical trials. However, the clinical efficacy may be limited by the disadvantages of vulnerability to enzymatic degradation and low immunogenicity of peptides. To further improve the potency of vaccine, we developed a poly(lactide-co-glycolide)-acid (PLGA) nanoparticle, which encapsulated the antigenic peptide HPV16 E744-62, and used adenosine triphosphate (ATP), one of the most important intracellular metabolites and an endogenous extracellular danger signal for the immune system, as a new adjuvant component. The results showed that PLGA nanoparticles increased the in vivo stability, lymph node accumulation, and dendritic cell (DC) uptake of the E7 peptide; in addition, ATP further increased the migration, nanoparticle uptake, and maturation of DCs. Preventive immunization with ATP-adjuvanted nanoparticles completely abolished the growth of TC-1 tumors in mice and produced long-lasting immunity against tumor rechallenge. When tumors were fully established, therapeutic immunization with ATP-adjuvanted nanoparticles still significantly inhibited tumor progression. Mechanistically, ATP-adjuvanted nanoparticles significantly improved the systemic generation of antitumor effector cells, boosted the local functional status of these cells in tumors, and suppressed the generation and tumor infiltration of immunosuppressive Treg cells and myeloid-derived suppressor cells. These findings indicate that ATP is an effective vaccine adjuvant and that nanoparticles adjuvanted with ATP were able to elicit robust antitumor cellular immunity, which may provide a promising therapeutic vaccine candidate for the treatment of clinical malignancies, such as cervical cancer.
Collapse
Affiliation(s)
- Qishu Zhang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Weiwei Huang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Mingcui Yuan
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Weiran Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Liangqun Hua
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
- School of Life Science, Yunnan University, Kunming 650500, China
| | - Zhongqian Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Fulan Gao
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Sijin Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Chao Ye
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Yongjun Chen
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Jinrong He
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Wenjia Sun
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Xu Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Hongmei Bai
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Yanbing Ma
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| |
Collapse
|
104
|
Essential role of submandibular lymph node dendritic cells in protective sublingual immunotherapy against murine allergy. Commun Biol 2020; 3:742. [PMID: 33288832 PMCID: PMC7721894 DOI: 10.1038/s42003-020-01466-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 11/04/2020] [Indexed: 01/12/2023] Open
Abstract
While sublingual immunotherapy (SLIT) is known as an allergen-specific treatment for type-1 allergies, how it controls allergic pathogenesis remains unclear. Here, we show the prerequisite role of conventional dendritic cells in submandibular lymph nodes (ManLNs) in the effectiveness of SLIT for the treatment of allergic disorders in mice. Deficiency of conventional dendritic cells or CD4+Foxp3+ regulatory T (Treg) cells abrogates the protective effect of SLIT against allergic disorders. Furthermore, sublingual antigenic application primarily induces antigen-specific CD4+Foxp3+ Treg cells in draining ManLNs, in which it is severely impaired in the absence of cDCs. In ManLNs, migratory CD11b+ cDCs are superior to other conventional dendritic cell subsets for the generation of antigen-specific CD4+Foxp3+ Treg cells, which is reflected by their dominancy in the tolerogenic features to favor this program. Thus, ManLNs are privileged sites in triggering mucosal tolerance mediating protect effect of SLIT on allergic disorders that requires a tolerogenesis of migratory CD11b+ conventional dendritic cells. Noriaki Miyanaga and Hideaki Takagi et al. identify an essential role for migratory dendritic cells in mediating immunotherapy treatment against allergies in mice. They show that submandibular lymph node dendritic cells induce regulatory T cells, and their absence abrogates the effectiveness of immunotherapy treatment.
Collapse
|
105
|
Falode JA, Akinmoladun AC, Olaleye MT, Akindahunsi AA. Kigelia africana (Lam.) Benth leaf extract inhibits rat brain and liver mitochondrial membrane permeability transition pore opening. Drug Chem Toxicol 2020; 45:1614-1624. [PMID: 33280443 DOI: 10.1080/01480545.2020.1850757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The effect of Kigelia africana on mitochondrial membrane permeability transition has not been explored. In this study, the effect of a solvent fraction of Kigelia africana leaf extract on mitochondrial membrane permeability transition of rat brain and liver was evaluated. A methanol extract of K. africana leaves was fractionated into different solvents by vacuum liquid chromatography and following preliminary screening, the dichloromethane:ethylacetate (1:1) fraction was selected for further assays. Constituent phytochemicals in the fraction were revealed by thin-layer chromatography and further purification was carried out to characterize the compounds. Brain and liver mitochondria were isolated and used for mitochondrial membrane permeability transition and adenosine triphosphatase assays. Exogenous Ca2+ and Al3+ were used to trigger the mitochondrial membrane permeability transition opening. Physicochemical properties revealed by thin-layer chromatography showed that the isolated compounds were flavonoids. The extract inhibited mitochondrial membrane permeability transition opening in the presence and absence of triggering agents in brain and liver mitochondria. It also inhibited mitochondrial lipid peroxidation and adenosine triphosphatase activity. These results suggest that the extract can limit the rate of apoptosis via inhibition of mitochondrial membrane permeability transition which is pivotal to the mitochondrial apoptotic pathway and is an important therapeutic target in some pathological conditions.
Collapse
Affiliation(s)
- John Adeolu Falode
- Phytomedicine, Biochemical Pharmacology and Toxicology Unit, Department of Biochemistry, Federal University, Oye-Ekiti, Nigeria.,Phytomedicine, Biochemical Pharmacology and Toxicology Unit, Department of Biochemistry, School of Sciences, The Federal University of Technology, Akure, Nigeria
| | - Afolabi Clement Akinmoladun
- Phytomedicine, Biochemical Pharmacology and Toxicology Unit, Department of Biochemistry, School of Sciences, The Federal University of Technology, Akure, Nigeria
| | - Mary Tolulope Olaleye
- Phytomedicine, Biochemical Pharmacology and Toxicology Unit, Department of Biochemistry, School of Sciences, The Federal University of Technology, Akure, Nigeria
| | - Afolabi Akintunde Akindahunsi
- Phytomedicine, Biochemical Pharmacology and Toxicology Unit, Department of Biochemistry, School of Sciences, The Federal University of Technology, Akure, Nigeria
| |
Collapse
|
106
|
Cohen M, Giladi A, Raposo C, Zada M, Li B, Ruckh J, Deczkowska A, Mohar B, Shechter R, Lichtenstein RG, Amit I, Schwartz M. Meningeal lymphoid structures are activated under acute and chronic spinal cord pathologies. Life Sci Alliance 2020; 4:4/1/e202000907. [PMID: 33277355 PMCID: PMC7723261 DOI: 10.26508/lsa.202000907] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 12/01/2022] Open
Abstract
We found that acute insult to the central nervous system induces the formation of lymphocyte aggregates reminiscent of tertiary lymphoid structures within the spinal cord meninges. Unlike draining CNS-cervical lymph nodes, meningeal lymphocytes are locally activated during neuro-inflammtion and neurodegeneration. Tertiary lymphoid structures (TLS) are organized aggregates of B and T cells formed ectopically during different stages of life in response to inflammation, infection, or cancer. Here, we describe formation of structures reminiscent of TLS in the spinal cord meninges under several central nervous system (CNS) pathologies. After acute spinal cord injury, B and T lymphocytes locally aggregate within the meninges to form TLS-like structures, and continue to accumulate during the late phase of the response to the injury, with a negative impact on subsequent pathological conditions, such as experimental autoimmune encephalomyelitis. Using a chronic model of spinal cord pathology, the mSOD1 mouse model of amyotrophic lateral sclerosis, we further showed by single-cell RNA-sequencing that a meningeal lymphocyte niche forms, with a unique organization and activation state, including accumulation of pre-B cells in the spinal cord meninges. Such a response was not found in the CNS-draining cervical lymph nodes. The present findings suggest that a special immune response develops in the meninges during various neurological pathologies in the CNS, a possible reflection of its immune privileged nature.
Collapse
Affiliation(s)
- Merav Cohen
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Amir Giladi
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Catarina Raposo
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Mor Zada
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Baoguo Li
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Julia Ruckh
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Boaz Mohar
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Ravid Shechter
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Rachel G Lichtenstein
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Schwartz
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel .,Klarman Cell Observatory, Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| |
Collapse
|
107
|
Tabachnick-Cherny S, Pinto S, Berko D, Curato C, Wolf Y, Porat Z, Karmona R, Tirosh B, Jung S, Navon A. Polyglutamine-Related Aggregates Can Serve as a Potent Antigen Source for Cross-Presentation by Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2020; 205:2583-2594. [PMID: 33067378 DOI: 10.4049/jimmunol.1901535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 09/15/2020] [Indexed: 01/16/2023]
Abstract
Protective MHC class I-dependent immune responses require an overlap between repertoires of proteins directly presented on target cells and cross-presented by professional APC, specifically dendritic cells. How stable proteins that rely on defective ribosomal proteins for direct presentation are captured for cell-to-cell transfer remains enigmatic. In this study, we address this issue using a combination of in vitro (C57BL/6-derived mouse cell lines) and in vivo (C57BL/6 mouse strains) approaches involving stable and unstable versions of OVA model Ags displaying defective ribosomal protein-dependent and -independent Ag presentation, respectively. Apoptosis, but not necrosis, of donor cells was found associated with robust global protein aggregate formation and captured stable proteins permissive for cross-presentation. Potency of aggregates to serve as Ag source was directly demonstrated using polyglutamine-equipped model substrates. Collectively, our data implicate global protein aggregation in apoptotic cells as a mechanism that ensures the overlap between MHC class I epitopes presented directly or cross-presented by APC and demonstrate the unusual ability of dendritic cells to process stable protein aggregates.
Collapse
Affiliation(s)
- Shira Tabachnick-Cherny
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Sivan Pinto
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Dikla Berko
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Caterina Curato
- Department of Immunology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yochai Wolf
- Department of Immunology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ziv Porat
- Department of Biological Services, The Weizmann Institute of Science, Rehovot 76100, Israel; and
| | - Rotem Karmona
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Boaz Tirosh
- The Institute for Drug Research, The School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Steffen Jung
- Department of Immunology, The Weizmann Institute of Science, Rehovot 76100, Israel;
| | - Ami Navon
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel;
| |
Collapse
|
108
|
Que W, Guo WZ, Li XK. Manipulation of Regulatory Dendritic Cells for Induction Transplantation Tolerance. Front Immunol 2020; 11:582658. [PMID: 33162996 PMCID: PMC7591396 DOI: 10.3389/fimmu.2020.582658] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 08/31/2020] [Indexed: 12/13/2022] Open
Abstract
Current organ transplantation therapy is life-saving but accompanied by well-recognized side effects due to post-transplantation systematic immunosuppressive treatment. Dendritic cells (DCs) are central instigators and regulators of transplantation immunity and are responsible for balancing allograft rejection and tolerance. They are derived from monocyte-macrophage DC progenitors originating in the bone marrow and are classified into different subsets based on their developmental, phenotypical, and functional criteria. Functionally, DCs instigate allograft immunity by presenting donor antigens to alloreactive T cells via direct, indirect, and semidirect recognition pathways and provide essential signaling for alloreactive T cell activation via costimulatory molecules and pro-inflammatory cytokines. Regulatory DCs (DCregs) are characterized by a relatively low expression of major histocompatibility complex, costimulatory molecules, and altered cytokine production and exert their regulatory function through T cell anergy, T cell deletion, and regulatory T cell induction. In rodent transplantation studies, DCreg-based therapy, by in situ targeting or infusion of ex vivo generated DCregs, exhibits promising potential as a natural, well-tolerated, organ-specific therapeutic strategy for promoting lasting organ-specific transplantation tolerance. Recent early-phase studies of DCregs have begun to examine the safety and efficacy of DCreg-induced allograft tolerance in living-donor renal or liver transplantations. The present review summarizes the basic characteristics, function, and translation of DCregs in transplantation tolerance induction.
Collapse
Affiliation(s)
- Weitao Que
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Wen-Zhi Guo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiao-Kang Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| |
Collapse
|
109
|
Abuga KM, Jones-Warner W, Hafalla JCR. Immune responses to malaria pre-erythrocytic stages: Implications for vaccine development. Parasite Immunol 2020; 43:e12795. [PMID: 32981095 PMCID: PMC7612353 DOI: 10.1111/pim.12795] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 08/26/2020] [Accepted: 09/14/2020] [Indexed: 12/16/2022]
Abstract
Radiation-attenuated sporozoites induce sterilizing immunity and remain the 'gold standard' for malaria vaccine development. Despite practical challenges in translating these whole sporozoite vaccines to large-scale intervention programmes, they have provided an excellent platform to dissect the immune responses to malaria pre-erythrocytic (PE) stages, comprising both sporozoites and exoerythrocytic forms. Investigations in rodent models have provided insights that led to the clinical translation of various vaccine candidates-including RTS,S/AS01, the most advanced candidate currently in a trial implementation programme in three African countries. With advances in immunology, transcriptomics and proteomics, and application of lessons from past failures, an effective, long-lasting and wide-scale malaria PE vaccine remains feasible. This review underscores the progress in PE vaccine development, focusing on our understanding of host-parasite immunological crosstalk in the tissue environments of the skin and the liver. We highlight possible gaps in the current knowledge of PE immunity that can impact future malaria vaccine development efforts.
Collapse
Affiliation(s)
- Kelvin Mokaya Abuga
- Department of Infection Biology, Faculty of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, UK.,Department of Epidemiology and Demography, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - William Jones-Warner
- Department of Infection Biology, Faculty of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Julius Clemence R Hafalla
- Department of Infection Biology, Faculty of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
110
|
Tsukamoto H, Kouwaki T, Oshiumi H. Aging-Associated Extracellular Vesicles Contain Immune Regulatory microRNAs Alleviating Hyperinflammatory State and Immune Dysfunction in the Elderly. iScience 2020; 23:101520. [PMID: 32927264 PMCID: PMC7495115 DOI: 10.1016/j.isci.2020.101520] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/30/2020] [Accepted: 08/27/2020] [Indexed: 12/16/2022] Open
Abstract
Aging-associated changes in the immune system often lead to immune dysfunction; however, the mechanisms that underlie this phenomenon have yet to be fully elucidated. This study found that the microRNA-192 (miR-192) is an aging-associated immune regulatory microRNA whose concentration was significantly increased in aged extracellular vesicles (EVs) due to the hyperinflammatory state of aged mice. Interestingly, EV miR-192 exhibited anti-inflammatory effects on macrophages. In our aged mouse model, aging was associated with prolonged inflammation in the lung upon stimulation with inactivated influenza whole virus particles (WVP), whereas EV miR-192 alleviated the prolonged inflammation associated with aging. The hyperinflammatory state of aged mice resulted in reduced production of specific antibodies and efficacy of vaccination with WVP; however, EV miR-192 attenuated this hyperinflammatory state and improved vaccination efficacy in aged mice. Our data indicate that aged EVs constitute a negative feedback loop that alleviates aging-associated immune dysfunction.
Collapse
Affiliation(s)
- Hirotake Tsukamoto
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Takahisa Kouwaki
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Hiroyuki Oshiumi
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| |
Collapse
|
111
|
Huang MN, Nicholson LT, Batich KA, Swartz AM, Kopin D, Wellford S, Prabhakar VK, Woroniecka K, Nair SK, Fecci PE, Sampson JH, Gunn MD. Antigen-loaded monocyte administration induces potent therapeutic antitumor T cell responses. J Clin Invest 2020; 130:774-788. [PMID: 31661470 DOI: 10.1172/jci128267] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 10/22/2019] [Indexed: 12/20/2022] Open
Abstract
Efficacy of dendritic cell (DC) cancer vaccines is classically thought to depend on their antigen-presenting cell (APC) activity. Studies show, however, that DC vaccine priming of cytotoxic T lymphocytes (CTLs) requires the activity of endogenous DCs, suggesting that exogenous DCs stimulate antitumor immunity by transferring antigens (Ags) to endogenous DCs. Such Ag transfer functions are most commonly ascribed to monocytes, implying that undifferentiated monocytes would function equally well as a vaccine modality and need not be differentiated to DCs to be effective. Here, we used several murine cancer models to test the antitumor efficacy of undifferentiated monocytes loaded with protein or peptide Ag. Intravenously injected monocytes displayed antitumor activity superior to DC vaccines in several cancer models, including aggressive intracranial glioblastoma. Ag-loaded monocytes induced robust CTL responses via Ag transfer to splenic CD8+ DCs in a manner independent of monocyte APC activity. Ag transfer required cell-cell contact and the formation of connexin 43-containing gap junctions between monocytes and DCs. These findings demonstrate the existence of an efficient gap junction-mediated Ag transfer pathway between monocytes and CD8+ DCs and suggest that administration of tumor Ag-loaded undifferentiated monocytes may serve as a simple and efficacious immunotherapy for the treatment of human cancers.
Collapse
Affiliation(s)
- Min-Nung Huang
- Department of Immunology.,Division of Cardiology, Department of Medicine
| | | | - Kristen A Batich
- School of Medicine.,Department of Pathology.,Preston Robert Tisch Brain Tumor Center
| | - Adam M Swartz
- Department of Pathology.,Preston Robert Tisch Brain Tumor Center
| | | | | | | | - Karolina Woroniecka
- School of Medicine.,Department of Pathology.,Preston Robert Tisch Brain Tumor Center
| | - Smita K Nair
- Department of Pathology.,Preston Robert Tisch Brain Tumor Center.,Department of Neurosurgery, and.,Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Peter E Fecci
- Department of Pathology.,Preston Robert Tisch Brain Tumor Center.,Department of Neurosurgery, and
| | - John H Sampson
- Department of Pathology.,Preston Robert Tisch Brain Tumor Center.,Department of Neurosurgery, and
| | - Michael D Gunn
- Department of Immunology.,Division of Cardiology, Department of Medicine
| |
Collapse
|
112
|
Sun T, Nguyen A, Gommerman JL. Dendritic Cell Subsets in Intestinal Immunity and Inflammation. THE JOURNAL OF IMMUNOLOGY 2020; 204:1075-1083. [PMID: 32071090 DOI: 10.4049/jimmunol.1900710] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 10/11/2019] [Indexed: 12/21/2022]
Abstract
The mammalian intestine is a complex environment that is constantly exposed to Ags derived from food, microbiota, and metabolites. Intestinal dendritic cells (DC) have the responsibility of establishing oral tolerance against these Ags while initiating immune responses against mucosal pathogens. We now know that DC are a heterogeneous population of innate immune cells composed of classical and monocyte-derived DC, Langerhans cells, and plasmacytoid DC. In the intestine, DC are found in organized lymphoid tissues, such as the mesenteric lymph nodes and Peyer's patches, as well as in the lamina propria. In this Brief Review, we review recent work that describes a division of labor between and collaboration among gut DC subsets in the context of intestinal homeostasis and inflammation. Understanding relationships between DC subtypes and their biological functions will rationalize oral vaccine design and will provide insights into treatments that quiet pathological intestinal inflammation.
Collapse
Affiliation(s)
- Tian Sun
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S1A8, Canada
| | - Albert Nguyen
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S1A8, Canada
| | - Jennifer L Gommerman
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S1A8, Canada
| |
Collapse
|
113
|
Liu Z, He J, Zhu T, Hu C, Bo R, Wusiman A, Hu Y, Wang D. Lentinan-Functionalized Graphene Oxide Is an Effective Antigen Delivery System That Modulates Innate Immunity and Improves Adaptive Immunity. ACS APPLIED MATERIALS & INTERFACES 2020; 12:39014-39023. [PMID: 32805921 DOI: 10.1021/acsami.0c12078] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Graphene oxide (GO) and lentinan have received great attention because of their utility in biomedical applications. Graphene oxide is utilized in drug- and vaccine-delivery systems due to its biocompatibility, large surface area, and outstanding adsorption capability, while lentinan has immunity-enhancing effects. In this study, we synthesized and characterized GO grafted with lentinan (LNT) as an adjuvant and investigated how to impact the immune responses. Lentinan-modified GO (GO-LNT) facilitated antigen uptake in macrophages and improved the efficiency of antigen application in vitro. Furthermore, in vivo, compared with GO/OVA, GO-LNT/OVA decreased the release rate of ovalbumin (OVA) to sustain long-term immune responses and boost the levels of IgG and IgG subtypes. Hence, we can infer that the effects of GO-LNT were a result of the increased amounts of antigen uptake by cells. Overall, our studies demonstrated that GO-LNT could suffice for a safe and effective vaccine-delivery system as well as an excellent adjuvant that both elicits a long-term immune memory response and potentiates cellular and humoral immunity.
Collapse
Affiliation(s)
- Zhenguang Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Jin He
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Tianyu Zhu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Cong Hu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Ruonan Bo
- School of Veterinary Medicine, Yangzhou University, Yangzhou 225009, P. R. China
| | - Adelijiang Wusiman
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Yuanliang Hu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Deyun Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| |
Collapse
|
114
|
Kerr MD, McBride DA, Chumber AK, Shah NJ. Combining therapeutic vaccines with chemo- and immunotherapies in the treatment of cancer. Expert Opin Drug Discov 2020; 16:89-99. [PMID: 32867561 DOI: 10.1080/17460441.2020.1811673] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Breakthroughs in cancer immunotherapy have spurred interest in the development of vaccines to mediate prophylactic protection and therapeutic efficacy against primary tumors or to prevent relapse. However, immunosuppressive mechanisms employed by cancer cells to generate effective resistance have hampered clinical translation of therapeutic cancer vaccines. To enhance vaccine efficacy, the immunomodulatory properties of cytoreductive therapies could amplify a cancer-specific immune response. AREAS COVERED Herein, the authors discuss therapeutic cancer vaccines that harness whole cells and antigen-targeted vaccines. First, recent advancements in both autologous and allogeneic whole-cell vaccines and combinations with checkpoint blockade and chemotherapy are reviewed. Next, tumor antigen-targeted vaccines using peptide-based vaccines and DNA-vaccines are discussed. Finally, combination therapies using antigen-targeted vaccines are reviewed. EXPERT OPINION A deeper understanding of the immunostimulatory properties of cytoreductive therapies has supported their utility in combination therapies involving cancer vaccines as a potential strategy to induce a durable anti-tumor immune response for multiple types of cancers. Based on current evidence, combination therapies may have synergies that depend on the identity of the cytotoxic agent, vaccine target, dosing schedule, and cancer type. Together, these observations suggest that combining cancer vaccines with immunomodulatory cytoreductive therapy is a promising strategy for cancer therapy.
Collapse
Affiliation(s)
- Matthew D Kerr
- Department of NanoEngineering, University of California , San Diego, CA, USA.,Program in Chemical Engineering, University of California , San Diego, CA, USA.,Center for Nano-Immuno Engineering, University of California , San Diego, CA, USA
| | - David A McBride
- Department of NanoEngineering, University of California , San Diego, CA, USA.,Program in Chemical Engineering, University of California , San Diego, CA, USA.,Center for Nano-Immuno Engineering, University of California , San Diego, CA, USA
| | - Arun K Chumber
- Department of NanoEngineering, University of California , San Diego, CA, USA.,Program in Chemical Engineering, University of California , San Diego, CA, USA.,Center for Nano-Immuno Engineering, University of California , San Diego, CA, USA
| | - Nisarg J Shah
- Department of NanoEngineering, University of California , San Diego, CA, USA.,Program in Chemical Engineering, University of California , San Diego, CA, USA.,Center for Nano-Immuno Engineering, University of California , San Diego, CA, USA.,Program in Immunology, University of California , San Diego, CA, USA.,San Diego Center for Precision Immunotherapy, Moores Cancer Center, University of California , San Diego, CA, USA
| |
Collapse
|
115
|
Wei JJ, Kim HS, Spencer CA, Brennan-Crispi D, Zheng Y, Johnson NM, Rosenbach M, Miller C, Leung DH, Cotsarelis G, Leung TH. Activation of TRPA1 nociceptor promotes systemic adult mammalian skin regeneration. Sci Immunol 2020; 5:5/50/eaba5683. [PMID: 32859683 DOI: 10.1126/sciimmunol.aba5683] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/26/2020] [Accepted: 08/05/2020] [Indexed: 12/24/2022]
Abstract
Adult mammalian wounds, with rare exception, heal with fibrotic scars that severely disrupt tissue architecture and function. Regenerative medicine seeks methods to avoid scar formation and restore the original tissue structures. We show in three adult mouse models that pharmacologic activation of the nociceptor TRPA1 on cutaneous sensory neurons reduces scar formation and can also promote tissue regeneration. Local activation of TRPA1 induces tissue regeneration on distant untreated areas of injury, demonstrating a systemic effect. Activated TRPA1 stimulates local production of interleukin-23 (IL-23) by dermal dendritic cells, leading to activation of circulating dermal IL-17-producing γδ T cells. Genetic ablation of TRPA1, IL-23, dermal dendritic cells, or γδ T cells prevents TRPA1-mediated tissue regeneration. These results reveal a cutaneous neuroimmune-regeneration cascade triggered by topical TRPA1 activators that promotes adult mammalian tissue regeneration, presenting a new avenue for research and development of therapies for wounds and scars.
Collapse
Affiliation(s)
- Jenny J Wei
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Hali S Kim
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Casey A Spencer
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Donna Brennan-Crispi
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Ying Zheng
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Nicolette M Johnson
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Misha Rosenbach
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Christopher Miller
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Denis H Leung
- Singapore Management University, Singapore, Singapore
| | - George Cotsarelis
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA.,Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas H Leung
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA. .,Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
| |
Collapse
|
116
|
Zhou M, Sacirbegovic F, Zhao K, Rosenberger S, Shlomchik WD. T cell exhaustion and a failure in antigen presentation drive resistance to the graft-versus-leukemia effect. Nat Commun 2020; 11:4227. [PMID: 32839441 PMCID: PMC7445289 DOI: 10.1038/s41467-020-17991-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 07/28/2020] [Indexed: 12/20/2022] Open
Abstract
In hematopoietic cell transplants, alloreactive T cells mediate the graft-versus-leukemia (GVL) effect. However, leukemia relapse accounts for nearly half of deaths. Understanding GVL failure requires a system in which GVL-inducing T cells can be tracked. We used such a model wherein GVL is exclusively mediated by T cells that recognize the minor histocompatibility antigen H60. Here we report that GVL fails due to insufficient H60 presentation and T cell exhaustion. Leukemia-derived H60 is inefficiently cross-presented whereas direct T cell recognition of leukemia cells intensifies exhaustion. The anti-H60 response is augmented by H60-vaccination, an agonist αCD40 antibody (FGK45), and leukemia apoptosis. T cell exhaustion is marked by inhibitory molecule upregulation and the development of TOX+ and CD39-TCF-1+ cells. PD-1 blockade diminishes exhaustion and improves GVL, while blockade of Tim-3, TIGIT or LAG3 is ineffective. Of all interventions, FGK45 administration at the time of transplant is the most effective at improving memory and naïve T cell anti-H60 responses and GVL. Our studies define important causes of GVL failure and suggest strategies to overcome them.
Collapse
Affiliation(s)
- Meng Zhou
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Faruk Sacirbegovic
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kai Zhao
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sarah Rosenberger
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Warren D Shlomchik
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- The Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- The Hillman UPMC Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
117
|
He J, Liu Z, Jiang W, Zhu T, Wusiman A, Gu P, Liu J, Wang D. Immune-adjuvant activity of lentinan-modified calcium carbonate microparticles on a H 5N 1 vaccine. Int J Biol Macromol 2020; 163:1384-1392. [PMID: 32758599 DOI: 10.1016/j.ijbiomac.2020.08.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/24/2020] [Accepted: 08/01/2020] [Indexed: 12/20/2022]
Abstract
In recent years, the high prevalence of avian influenza viruses especially H5N1 subtype isolated from poultry and human has become a major public health concern. Vaccination is still a major strategy for preventing H5N1 infections. Lentinan (LNT), a β-1,3-glucohexaose with β-1,6-branches, is extracted from Lentinus edodes and has been extensively studied for its immunoenhancement effects. In this study, we synthesized and characterized calcium carbonate (CaCO3) microparticles which modified with LNT as an adjuvant for H5N1 vaccine and investigated their ability to enhance immune responses. We prepared spherical and uniform CaCO3-LNT microparticles with a mean hydrodynamic size was around 2 μm. The H5N1 antigen-loaded CaCO3-LNT particles were injected into mice to evaluate their effectiveness as an adjuvant for H5N1 vaccines. The results demonstrated that CaCO3-LNT/H5N1 significantly enhanced the expression of MHC-II and CD86 in lymph node dendritic cells, and increased the ratio of CD4+ to CD8+ T cells in lymphocytes. Moreover, CaCO3-LNT/H5N1 surprisingly increased the HI titers and induced the secretion of IgG subtypes (IgG1 and IgG2b) and Th-associated cytokines (TNF-α, IFN-γ and IL-4) in immunized mice. Therefore, by combining with the immunostimulatory activity of LNT and the drug/antigen delivery capabilities of CaCO3, the CaCO3-LNT/H5N1 could induce a stronger cellular and humoral immune response and could be a potential adjuvant for the H5N1 vaccine.
Collapse
Affiliation(s)
- Jin He
- Institution of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 21005, PR China
| | - Zhenguang Liu
- Institution of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 21005, PR China
| | - Wenming Jiang
- China Animal Health and Epidemiology Center, Qingdao, PR China
| | - Tianyu Zhu
- Institution of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 21005, PR China
| | - Adelijiang Wusiman
- Institution of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 21005, PR China
| | - Pengfei Gu
- Institution of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 21005, PR China
| | - Jiaguo Liu
- Institution of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 21005, PR China
| | - Deyun Wang
- Institution of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 21005, PR China.
| |
Collapse
|
118
|
Hu F, Yue H, Lu T, Ma G. Cytosolic delivery of HBsAg and enhanced cellular immunity by pH-responsive liposome. J Control Release 2020; 324:460-470. [DOI: 10.1016/j.jconrel.2020.05.042] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/24/2020] [Accepted: 05/25/2020] [Indexed: 01/10/2023]
|
119
|
Blumenthal D, Chandra V, Avery L, Burkhardt JK. Mouse T cell priming is enhanced by maturation-dependent stiffening of the dendritic cell cortex. eLife 2020; 9:e55995. [PMID: 32720892 PMCID: PMC7417170 DOI: 10.7554/elife.55995] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 07/27/2020] [Indexed: 12/28/2022] Open
Abstract
T cell activation by dendritic cells (DCs) involves forces exerted by the T cell actin cytoskeleton, which are opposed by the cortical cytoskeleton of the interacting antigen-presenting cell. During an immune response, DCs undergo a maturation process that optimizes their ability to efficiently prime naïve T cells. Using atomic force microscopy, we find that during maturation, DC cortical stiffness increases via a process that involves actin polymerization. Using stimulatory hydrogels and DCs expressing mutant cytoskeletal proteins, we find that increasing stiffness lowers the agonist dose needed for T cell activation. CD4+ T cells exhibit much more profound stiffness dependency than CD8+ T cells. Finally, stiffness responses are most robust when T cells are stimulated with pMHC rather than anti-CD3ε, consistent with a mechanosensing mechanism involving receptor deformation. Taken together, our data reveal that maturation-associated cytoskeletal changes alter the biophysical properties of DCs, providing mechanical cues that costimulate T cell activation.
Collapse
Affiliation(s)
- Daniel Blumenthal
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Vidhi Chandra
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Lyndsay Avery
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Janis K Burkhardt
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
120
|
Kim K, Ji P, Song M, Che TM, Bravo D, Pettigrew JE, Liu Y. Dietary plant extracts modulate gene expression profiles in alveolar macrophages of pigs experimentally infected with porcine reproductive and respiratory syndrome virus. J Anim Sci Biotechnol 2020; 11:74. [PMID: 32685145 PMCID: PMC7359597 DOI: 10.1186/s40104-020-00475-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/17/2020] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Our previous study showed that 3 plant extracts enhanced the immune responses and growth efficiency of weaned pigs infected with porcine reproductive and respiratory syndrome virus (PRRSV), which is one of the most economically important disease in swine industry. However, each plant extract differently effected on growth efficiency and immune responses. Therefore, the objective of this study was conducted to characterize the effects and investigate the potential underlying mechanisms of 3 plant extracts on gene expression of alveolar macrophages in weaned pigs experimentally infected with PRRSV. RESULTS PRRSV infection altered (P < 0.05) the expression of 1,352 genes in pigs fed the control (CON; 755 up, 597 down). Compared with the infected CON, feeding capsicum (CAP), garlic botanical (GAR), or turmeric oleoresin (TUR) altered the expression of 46 genes (24 up, 22 down), 134 genes (59 up, 75 down), or 98 genes (55 up, 43 down) in alveolar macrophages of PRRSV-infected pigs, respectively. PRRSV infection up-regulated (P < 0.05) the expression of genes related to cell apoptosis, immune system process, and response to stimulus, but down-regulated (P < 0.05) the expression of genes involved in signaling transduction and innate immune response. Compared with the infected CON, feeding TUR or GAR reduced (P < 0.05) the expression of genes associated with antigen processing and presentation, feeding CAP up-regulated (P < 0.05) the expression of genes involved in antigen processing and presentation. Supplementation of CAP, GAR, or TUR also enhanced (P < 0.05) the expression of several genes related to amino acid metabolism, steroid hormone synthesis, or RNA degradation, respectively. CONCLUSIONS The results suggest that 3 plant extracts differently regulated the expression of genes in alveolar macrophages of PRRSV-infected pigs, especially altering genes involved in immunity.
Collapse
Affiliation(s)
- Kwangwook Kim
- Department of Animal Science, University of California, Davis, CA USA
| | - Peng Ji
- Department of Nutrition, University of California, Davis, CA USA
| | - Minho Song
- Department of Animal Science and Biotechnology, Chungnam National University, Daejeon, South Korea
| | - Tung M. Che
- Department of Animal Production, Nong Lam University, Ho Chi Minh City, Vietnam
| | - David Bravo
- Pancosma SA, Geneva, Switzerland
- Current address: Land O’Lakes Inc., Arden Hills, MN USA
| | | | - Yanhong Liu
- Department of Animal Science, University of California, Davis, CA USA
| |
Collapse
|
121
|
Allen BM, Hiam KJ, Burnett CE, Venida A, DeBarge R, Tenvooren I, Marquez DM, Cho NW, Carmi Y, Spitzer MH. Systemic dysfunction and plasticity of the immune macroenvironment in cancer models. Nat Med 2020; 26:1125-1134. [PMID: 32451499 PMCID: PMC7384250 DOI: 10.1038/s41591-020-0892-6] [Citation(s) in RCA: 187] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 04/17/2020] [Indexed: 02/07/2023]
Abstract
Understanding of the factors governing immune responses in cancer remains incomplete, limiting patient benefit. In this study, we used mass cytometry to define the systemic immune landscape in response to tumor development across five tissues in eight mouse tumor models. Systemic immunity was dramatically altered across models and time, with consistent findings in the peripheral blood of patients with breast cancer. Changes in peripheral tissues differed from those in the tumor microenvironment. Mice with tumor-experienced immune systems mounted dampened responses to orthogonal challenges, including reduced T cell activation during viral or bacterial infection. Antigen-presenting cells (APCs) mounted weaker responses in this context, whereas promoting APC activation rescued T cell activity. Systemic immune changes were reversed with surgical tumor resection, and many were prevented by interleukin-1 or granulocyte colony-stimulating factor blockade, revealing remarkable plasticity in the systemic immune state. These results demonstrate that tumor development dynamically reshapes the composition and function of the immune macroenvironment.
Collapse
Affiliation(s)
- Breanna M Allen
- Graduate Program in Biomedical Sciences, University of California, San Francisco, San Francisco, CA, USA
- Departments of Otolaryngology and Microbiology & Immunology, Helen Diller Family Comprehensive Cancer Center, Parker Institute for Cancer Immunotherapy, Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, CA, USA
| | - Kamir J Hiam
- Graduate Program in Biomedical Sciences, University of California, San Francisco, San Francisco, CA, USA
- Departments of Otolaryngology and Microbiology & Immunology, Helen Diller Family Comprehensive Cancer Center, Parker Institute for Cancer Immunotherapy, Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, CA, USA
| | - Cassandra E Burnett
- Graduate Program in Biomedical Sciences, University of California, San Francisco, San Francisco, CA, USA
- Departments of Otolaryngology and Microbiology & Immunology, Helen Diller Family Comprehensive Cancer Center, Parker Institute for Cancer Immunotherapy, Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, CA, USA
| | - Anthony Venida
- Graduate Program in Biomedical Sciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
| | - Rachel DeBarge
- Graduate Program in Biomedical Sciences, University of California, San Francisco, San Francisco, CA, USA
- Departments of Otolaryngology and Microbiology & Immunology, Helen Diller Family Comprehensive Cancer Center, Parker Institute for Cancer Immunotherapy, Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, CA, USA
| | - Iliana Tenvooren
- Departments of Otolaryngology and Microbiology & Immunology, Helen Diller Family Comprehensive Cancer Center, Parker Institute for Cancer Immunotherapy, Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, CA, USA
| | - Diana M Marquez
- Departments of Otolaryngology and Microbiology & Immunology, Helen Diller Family Comprehensive Cancer Center, Parker Institute for Cancer Immunotherapy, Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, CA, USA
| | - Nam Woo Cho
- Departments of Otolaryngology and Microbiology & Immunology, Helen Diller Family Comprehensive Cancer Center, Parker Institute for Cancer Immunotherapy, Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, CA, USA
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Yaron Carmi
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Matthew H Spitzer
- Graduate Program in Biomedical Sciences, University of California, San Francisco, San Francisco, CA, USA.
- Departments of Otolaryngology and Microbiology & Immunology, Helen Diller Family Comprehensive Cancer Center, Parker Institute for Cancer Immunotherapy, Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
122
|
Masuda T, Amann L, Sankowski R, Staszewski O, Lenz M, D Errico P, Snaidero N, Costa Jordão MJ, Böttcher C, Kierdorf K, Jung S, Priller J, Misgeld T, Vlachos A, Meyer-Luehmann M, Knobeloch KP, Prinz M. Novel Hexb-based tools for studying microglia in the CNS. Nat Immunol 2020; 21:802-815. [PMID: 32541832 DOI: 10.1038/s41590-020-0707-4] [Citation(s) in RCA: 186] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 05/07/2020] [Indexed: 12/26/2022]
Abstract
Microglia and central nervous system (CNS)-associated macrophages (CAMs), such as perivascular and meningeal macrophages, are implicated in virtually all diseases of the CNS. However, little is known about their cell-type-specific roles in the absence of suitable tools that would allow for functional discrimination between the ontogenetically closely related microglia and CAMs. To develop a new microglia gene targeting model, we first applied massively parallel single-cell analyses to compare microglia and CAM signatures during homeostasis and disease and identified hexosaminidase subunit beta (Hexb) as a stably expressed microglia core gene, whereas other microglia core genes were substantially downregulated during pathologies. Next, we generated HexbtdTomato mice to stably monitor microglia behavior in vivo. Finally, the Hexb locus was employed for tamoxifen-inducible Cre-mediated gene manipulation in microglia and for fate mapping of microglia but not CAMs. In sum, we provide valuable new genetic tools to specifically study microglia functions in the CNS.
Collapse
Affiliation(s)
- Takahiro Masuda
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.
| | - Lukas Amann
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Roman Sankowski
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Berta-Ottenstein-Programme for Clinician Scientists, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ori Staszewski
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Berta-Ottenstein-Programme for Clinician Scientists, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maximilian Lenz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Paolo D Errico
- Department of Neurology, Medical Center-University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nicolas Snaidero
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | | | - Chotima Böttcher
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Katrin Kierdorf
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Josef Priller
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
- DZNE and BIH, Berlin, Germany
- University of Edinburgh and UK DRI, Edinburgh, UK
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie Meyer-Luehmann
- Department of Neurology, Medical Center-University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Klaus-Peter Knobeloch
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
123
|
Abstract
PURPOSE OF REVIEW During the last decades, the field of regenerative medicine has been rapidly evolving. Major progress has been made in the development of biological substitutes applying the principles of cell transplantation, material science, and bioengineering. RECENT FINDINGS Among other sources, amniotic-derived products have been used for decades in various fields of medicine as a biomaterial for the wound care and tissue replacement. Moreover, human amniotic epithelial and mesenchymal cells have been intensively studied for their immunomodulatory capacities. Amniotic cells possess two major characteristics that have already been widely exploited. The first is their ability to modulate and suppress the innate and adaptive immunities, making them a true asset for chronic inflammatory disorders and for the induction of tolerance in transplantation models. The second is their multilineage differentiation capacity, offering a source of cells for tissue engineering. The latter combined with the use of amniotic membrane as a scaffold offers all components necessary to create an optimal environment for cell and tissue regeneration. This review summarizes beneficial properties of hAM and its derivatives and discusses their potential in regenerative medicine.
Collapse
Affiliation(s)
- Charles-Henri Wassmer
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Ekaterine Berishvili
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Institute of Medical Research, Ilia State University, Tbilisi, Georgia
| |
Collapse
|
124
|
Neoplastic fibrocytes play an essential role in bone marrow fibrosis in Jak2V617F-induced primary myelofibrosis mice. Leukemia 2020; 35:454-467. [PMID: 32472085 PMCID: PMC7862060 DOI: 10.1038/s41375-020-0880-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/07/2020] [Accepted: 05/18/2020] [Indexed: 11/23/2022]
Abstract
Primary myelofibrosis (PMF) is a myeloproliferative neoplasm (MPN) characterized by clonal myeloproliferation, progressive bone marrow (BM) fibrosis, splenomegaly, and anemia. BM fibrosis was previously thought to be a reactive phenomenon induced by mesenchymal stromal cells that are stimulated by the overproduction of cytokines such as transforming growth factor (TGF)-β1. However, the involvement of neoplastic fibrocytes in BM fibrosis was recently reported. In this study, we showed that the vast majority of collagen- and fibronectin-producing cells in the BM and spleens of Jak2V617F-induced myelofibrosis (MF) mice were fibrocytes derived from neoplastic hematopoietic cells. Neoplastic monocyte depletion eliminated collagen- and fibronectin-producing fibrocytes in BM and spleen, and ameliorated most characteristic MF features in Jak2V617F transgenic mice, including BM fibrosis, anemia, and splenomegaly, while had little effect on the elevated numbers of megakaryocytes and stem cells in BM, and leukothrombocytosis in peripheral blood. TGF-β1, which was produced by hematopoietic cells including fibrocytes, promoted the differentiation of neoplastic monocytes to fibrocytes, and elevated plasma TGF-β1 levels were normalized by monocyte depletion. Collectively, our data suggest that neoplastic fibrocytes are the major contributor to BM fibrosis in PMF, and TGF-β1 is required for their differentiation.
Collapse
|
125
|
Audiger C, Fois A, Thomas AL, Janssen E, Pelletier M, Lesage S. Merocytic Dendritic Cells Compose a Conventional Dendritic Cell Subset with Low Metabolic Activity. THE JOURNAL OF IMMUNOLOGY 2020; 205:121-132. [PMID: 32461238 DOI: 10.4049/jimmunol.1900970] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 04/29/2020] [Indexed: 12/14/2022]
Abstract
Conventional dendritic cells (cDCs) are arguably the most potent APCs that induce the activation of naive T cells in response to pathogens. In addition, at steady-state, cDCs help maintain immune tolerance. Two subsets of cDCs have been extensively characterized, namely cDC1 and cDC2, each contributing differently to immune responses. Recently, another dendritic cell (DC) subset, termed merocytic DCs (mcDCs), was defined. In contrast to both cDC1 and cDC2, mcDCs reverse T cell anergy, properties that could be exploited to potentiate cancer treatments. Yet, whether mcDCs represent an unconventional DC or a cDC subset remains to be defined. In this article, we further characterize mcDCs and find that they bear true characteristics of cDC subsets. Indeed, as for cDCs, mcDCs express the cDC-restricted transcription factor Zbtb46 and display very potent APC activity. In addition, mcDC population dynamics parallels that of cDC1 and cDC2 in both reconstitution kinetic studies and parabiotic mice. We next investigated their relatedness to cDC1 and cDC2 and demonstrate that mcDCs are not dependent on cDC1-related Irf8 and Batf3 transcription factors, are dependent on Irf4, a cDC2-specific transcription factor, and express a unique transcriptomic signature. Finally, we find that cDC1, cDC2, and mcDCs all present with different metabolic phenotypes, in which mcDCs exhibit the lowest glucose uptake activity and mcDC survival is the least affected by glycolysis inhibition. Defining the properties of mcDCs in mice may help identify a functionally equivalent subset in humans leading to the development of innovative cancer immunotherapies.
Collapse
Affiliation(s)
- Cindy Audiger
- Department of Immunology-Oncology, Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Adrien Fois
- Department of Immunology-Oncology, Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Alyssa L Thomas
- Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Edith Janssen
- Janssen Research and Development, Spring House, PA 19477
| | - Martin Pelletier
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, Quebec G1V 4G2, Canada; and.,Département de Microbiologie-Infectiologie et d'Immunologie, Université Laval, Quebec City, Quebec G1V 0A6, Canada
| | - Sylvie Lesage
- Department of Immunology-Oncology, Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada; .,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| |
Collapse
|
126
|
Taylor GA, Huang HI, Fee BE, Youssef N, Jewell ML, Cantillana V, Schoenborn AA, Rogala AR, Buckley AF, Feng CG, Vallance BA, Gulati AS, Hammer GE. Irgm1-deficiency leads to myeloid dysfunction in colon lamina propria and susceptibility to the intestinal pathogen Citrobacter rodentium. PLoS Pathog 2020; 16:e1008553. [PMID: 32453761 PMCID: PMC7274479 DOI: 10.1371/journal.ppat.1008553] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/05/2020] [Accepted: 04/15/2020] [Indexed: 01/09/2023] Open
Abstract
IRGM and its mouse orthologue Irgm1 are dynamin-like proteins that regulate vesicular remodeling, intracellular microbial killing, and pathogen immunity. IRGM dysfunction is linked to inflammatory bowel disease (IBD), and while it is thought that defective intracellular killing of microbes underscores IBD susceptibility, studies have yet to address how IRGM/Irgm1 regulates immunity to microbes relevant to intestinal inflammation. Here we find that loss of Irgm1 confers marked susceptibility to Citrobacter rodentium, a noninvasive intestinal pathogen that models inflammatory responses to intestinal bacteria. Irgm1-deficient mice fail to control C. rodentium outgrowth in the intestine, leading to systemic pathogen spread and host mortality. Surprisingly, susceptibility due to loss of Irgm1 function was not linked to defective intracellular killing of C. rodentium or exaggerated inflammation, but was instead linked to failure to remodel specific colon lamina propria (C-LP) myeloid cells that expand in response to C. rodentium infection and are essential for C. rodentium immunity. Defective immune remodeling was most striking in C-LP monocytes, which were successfully recruited to the infected C-LP, but subsequently underwent apoptosis. Apoptotic susceptibility was induced by C. rodentium infection and was specific to this setting of pathogen infection, and was not apparent in other settings of intestinal inflammation. These studies reveal a novel role for Irgm1 in host defense and suggest that deficiencies in survival and remodeling of C-LP myeloid cells that control inflammatory intestinal bacteria may underpin IBD pathogenesis linked to IRGM dysfunction.
Collapse
Affiliation(s)
- Gregory A. Taylor
- Geriatric Research, Education, and Clinical Center, VA Health Care Center, Durham, North Carolina, United States of America
- Departments of Medicine, Division of Geriatrics, and Center for the Study of Aging and Human Development, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail: (GAT); (GEH)
| | - Hsin-I Huang
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Brian E. Fee
- Geriatric Research, Education, and Clinical Center, VA Health Care Center, Durham, North Carolina, United States of America
| | - Nourhan Youssef
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Mark L. Jewell
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Viviana Cantillana
- Departments of Medicine, Division of Geriatrics, and Center for the Study of Aging and Human Development, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Alexi A. Schoenborn
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Allison R. Rogala
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Pediatrics, Division of Gastroenterology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Anne F. Buckley
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Carl G. Feng
- Department of Infectious Diseases and Immunology, University of Sydney, Sydney, NSW, Australia
| | - Bruce A. Vallance
- Department of Pediatrics, Division of Gastroenterology, BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Ajay S. Gulati
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Pediatrics, Division of Gastroenterology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Gianna E. Hammer
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail: (GAT); (GEH)
| |
Collapse
|
127
|
Depletion of CD11c+ dendritic cells in apolipoprotein E-deficient mice limits angiotensin II-induced abdominal aortic aneurysm formation and growth. Clin Sci (Lond) 2020; 133:2203-2215. [PMID: 31696215 DOI: 10.1042/cs20190924] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 01/30/2023]
Abstract
OBJECTIVE The role of chronic inflammation in abdominal aortic aneurysm (AAA) is controversial. CD11c+ antigen-presenting cells (APCs) (dendritic cells (DCs)) have been reported in human AAA samples but their role is unclear. The effect of conditional depletion of CD11c+ cells on experimental AAA was investigated in the angiotensin II (AngII)-infused apolipoprotein E-deficient (ApoE-/-) mouse model. APPROACH CD11c-diphtheria toxin (DT or D.tox) receptor (DTR), ovalbumin (OVA) fragment aa 140-386, and enhanced green fluorescent protein (eGFP)-ApoE-/- (CD11c.DOG.ApoE-/-) mice were generated and CD11c+ cell depletion achieved with D.tox injections (8 ng/g body weight, i.p., every-other-day). AAA formation and growth were assessed by measurement of supra-renal aortic (SRA) diameter in vivo by serial ultrasound and by morphometry assessment of harvested aortas at the end of the study. RESULTS Depletion of CD11c+ cells by administration of D.tox on alternative days was shown to reduce the maximum diameter of AAAs induced by 28 days AngII infusion compared with controls (D.tox, 1.58 ± 0.03 mm vs Vehicle control, 1.81 ± 0.06 mm, P<0.001). CD11c+ depletion commencing after AAA establishment by 14 days of AngII infusion, was also shown to lead to smaller AAAs than controls after a further 14 days (D.tox, 1.54 ± 0.04 mm vs Vehicle control, 1.80 ± 0.03 mm, P<0.001). Flow cytometry revealed significantly lower numbers of circulating CD44hi CD62Llo effector CD4 T cells, CD44hi CD62Llo effector CD8 T cells and B220+ B cells in CD11c+ cell-depleted mice versus controls. CD11c+ depletion attenuated SRA matrix degradation indicated by decreased neutrophil elastase activity (P=0.014), lower elastin degradation score (P=0.012) and higher collagen content (P=0.002). CONCLUSION CD11c+ cell-depletion inhibited experimental AAA development and growth associated with down-regulation of circulating effector T cells and attenuated matrix degradation. The findings suggest involvement of autoreactive immune cells in AAA pathogenesis.
Collapse
|
128
|
Kidney dendritic cells: fundamental biology and functional roles in health and disease. Nat Rev Nephrol 2020; 16:391-407. [PMID: 32372062 DOI: 10.1038/s41581-020-0272-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2020] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DCs) are chief inducers of adaptive immunity and regulate local inflammatory responses across the body. Together with macrophages, the other main type of mononuclear phagocyte, DCs constitute the most abundant component of the intrarenal immune system. This network of functionally specialized immune cells constantly surveys its microenvironment for signs of injury or infection, which trigger the initiation of an immune response. In the healthy kidney, DCs coordinate effective immune responses, for example, by recruiting neutrophils for bacterial clearance in pyelonephritis. The pro-inflammatory actions of DCs can, however, also contribute to tissue damage in various types of acute kidney injury and chronic glomerulonephritis, as DCs recruit and activate effector T cells, which release toxic mediators and maintain tubulointerstitial immune infiltrates. These actions are counterbalanced by DC subsets that promote the activation and maintenance of regulatory T cells to support resolution of the immune response and allow kidney repair. Several studies have investigated the multiple roles for DCs in kidney homeostasis and disease, but it has become clear that current tools and subset markers are not sufficient to accurately distinguish DCs from macrophages. Multidimensional transcriptomic analysis studies promise to improve mononuclear phagocyte classification and provide a clearer view of DC ontogeny and subsets.
Collapse
|
129
|
Jung HE, Lee HK. Host Protective Immune Responses against Influenza A Virus Infection. Viruses 2020; 12:v12050504. [PMID: 32375274 PMCID: PMC7291249 DOI: 10.3390/v12050504] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/27/2020] [Accepted: 04/30/2020] [Indexed: 12/31/2022] Open
Abstract
Influenza viruses cause infectious respiratory disease characterized by fever, myalgia, and congestion, ranging in severity from mild to life-threating. Although enormous efforts have aimed to prevent and treat influenza infections, seasonal and pandemic influenza outbreaks remain a major public health concern. This is largely because influenza viruses rapidly undergo genetic mutations that restrict the long-lasting efficacy of vaccine-induced immune responses and therapeutic regimens. In this review, we discuss the virological features of influenza A viruses and provide an overview of current knowledge of the innate sensing of invading influenza viruses and the protective immune responses in the host.
Collapse
Affiliation(s)
- Hi Eun Jung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
- Correspondence: (H.E.J.); (H.K.L.); Tel.: +82-42-350-4281 (H.K.L.)
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
- KAIST Institute for Health Science and Technology, KAIST, Daejeon 34141, Korea
- Correspondence: (H.E.J.); (H.K.L.); Tel.: +82-42-350-4281 (H.K.L.)
| |
Collapse
|
130
|
Ebelt ND, Zuniga E, Johnson BL, Diamond DJ, Manuel ER. 5-Azacytidine Potentiates Anti-tumor Immunity in a Model of Pancreatic Ductal Adenocarcinoma. Front Immunol 2020; 11:538. [PMID: 32296439 PMCID: PMC7136411 DOI: 10.3389/fimmu.2020.00538] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 03/09/2020] [Indexed: 12/26/2022] Open
Abstract
Tumors evolve a variety of mechanisms to escape immune detection while expressing tumor-promoting molecules that can be immunogenic. Here, we show that transposable elements (TE) and gene encoded, tumor-associated antigens (TAA), which can be both highly immunogenic and tumor-promoting, are significantly upregulated during the transition from pre-malignancy to malignancy in an inducible model of pancreatic ductal adenocarcinoma (PDAC). Coincident with the increased presence of TEs and TAAs was the downregulation of gene transcripts associated with antigen presentation, T cell recruitment and intrinsic anti-viral responses, suggesting a unique strategy employed by PDAC to possibly augment tumorigenesis while escaping detection by the immune system. In vitro treatment of mouse and human PDAC cell lines with the DNA methyltransferase inhibitor 5-azacytidine (Aza) resulted in augmented expression of transcripts for antigen presentation machinery and T cell chemokines. When immunocompetent mice implanted with PDAC were therapeutically treated with Aza, we observed significant tumor regression that was not observed in immunocompromised mice, implicating anti-tumor immunity as the principal mechanism of tumor growth control. Analysis of PDAC tumors, immediately following Aza treatment in immunocompetent mice, revealed a significantly greater infiltration of T cells and various innate immune subsets compared to control treatment, suggesting that Aza treatment enhances tumor immunogenicity. Thus, augmenting antigen presentation and T cell chemokine expression using DNA methyltransferase inhibitors could be leveraged to potentiate adaptive anti-tumor immune responses against PDAC.
Collapse
Affiliation(s)
- Nancy D. Ebelt
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, United States
| | - Edith Zuniga
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, United States
| | - Benjamin L. Johnson
- Department of Hematology and Hematopoietic Stem Cell Transplantation, City of Hope, Duarte, CA, United States
| | - Don J. Diamond
- Department of Hematology and Hematopoietic Stem Cell Transplantation, City of Hope, Duarte, CA, United States
| | - Edwin R. Manuel
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, United States
| |
Collapse
|
131
|
The primacy of gastrointestinal tract antigen-presenting cells in lethal graft-versus-host disease. Blood 2020; 134:2139-2148. [PMID: 31697827 DOI: 10.1182/blood.2019000823] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/24/2019] [Indexed: 12/26/2022] Open
Abstract
Allogeneic stem cell transplantation is a cornerstone of curative therapy for high-risk and/or advanced hematological malignancies but remains limited by graft-versus-host disease (GVHD). GVHD is initiated by the interaction between recipient antigen-presenting cells (APCs) and donor T cells, culminating in T-cell differentiation along pathogenic type-1 and type-17 paradigms at the expense of tolerogenic regulatory T-cell patterns. Type-1 and type-17 T cells secrete cytokines (eg, granulocyte-macrophage colony-stimulating factor and interferon-γ) critical to the cytokine storm that amplifies expansion of donor APCs and their alloantigen presentation. It has become increasingly clear that pathogenic donor T-cell differentiation is initiated by both professional recipient APCs (eg, dendritic cells [DCs]) and nonprofessional APCs (eg, epithelial and mesenchymal cells), particularly within the gastrointestinal (GI) tract. In the immediate peritransplantation period, these APCs are profoundly modified by pathogen-associated molecular pattern (PAMP)/damage-associated molecular pattern (DAMP) signals derived from conditioning and intestinal microbiota. Subsequently, donor DCs in the GI tract are activated by DAMP/PAMP signals in the colon that gain access to the lamina propria once the mucosal barrier mucosa is compromised by GVHD. This results in donor DC expansion and alloantigen presentation in the colon and subsequent migration into the mesenteric lymph nodes. Here, new donor T cells are primed, expanded, differentiated, and imprinted with gut-homing integrins permissive of migration into the damaged GI tract, resulting in the lethal feed-forward cascade of GVHD. These new insights into our understanding of the cellular and molecular factors initiating GVHD, both spatially and temporally, give rise to a number of logical therapeutic targets, focusing on the inhibition of APC function in the GI tract.
Collapse
|
132
|
Cousin C, Oberkampf M, Felix T, Rosenbaum P, Weil R, Fabrega S, Morante V, Negri D, Cara A, Dadaglio G, Leclerc C. Persistence of Integrase-Deficient Lentiviral Vectors Correlates with the Induction of STING-Independent CD8 + T Cell Responses. Cell Rep 2020; 26:1242-1257.e7. [PMID: 30699352 DOI: 10.1016/j.celrep.2019.01.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 11/22/2018] [Accepted: 01/07/2019] [Indexed: 12/23/2022] Open
Abstract
Lentiviruses are among the most promising viral vectors for in vivo gene delivery. To overcome the risk of insertional mutagenesis, integrase-deficient lentiviral vectors (IDLVs) have been developed. We show here that strong and persistent specific cytotoxic T cell (CTL) responses are induced by IDLVs, which persist several months after a single injection. These responses were associated with the induction of mild and transient maturation of dendritic cells (DCs) and with the production of low levels of inflammatory cytokines and chemokines. They were independent of the IFN-I, TLR/MyD88, interferon regulatory factor (IRF), retinoic acid induced gene I (RIG-I), and stimulator of interferon genes (STING) pathways but require NF-κB signaling in CD11c+ DCs. Despite the lack of integration of IDLVs, the transgene persists for 3 months in the spleen and liver of IDLV-injected mice. These results demonstrate that the capacity of IDLVs to trigger persistent adaptive responses is mediated by a weak and transient innate response, along with the persistence of the vector in tissues.
Collapse
Affiliation(s)
- Céline Cousin
- Institut Pasteur, Unité de Régulation Immunitaire et Vaccinologie, Equipe Labellisée Ligue Contre le Cancer, 75015 Paris, France; INSERM U1041, 75015 Paris, France
| | - Marine Oberkampf
- Institut Pasteur, Unité de Régulation Immunitaire et Vaccinologie, Equipe Labellisée Ligue Contre le Cancer, 75015 Paris, France; INSERM U1041, 75015 Paris, France
| | - Tristan Felix
- Institut Pasteur, Unité de Régulation Immunitaire et Vaccinologie, Equipe Labellisée Ligue Contre le Cancer, 75015 Paris, France; INSERM U1041, 75015 Paris, France
| | - Pierre Rosenbaum
- Institut Pasteur, Unité de Régulation Immunitaire et Vaccinologie, Equipe Labellisée Ligue Contre le Cancer, 75015 Paris, France; INSERM U1041, 75015 Paris, France
| | - Robert Weil
- Institut Pasteur, Unité Signalisation et Pathogénèse, Département Biologie Cellulaire et Infection, 75015 Paris, France
| | - Sylvie Fabrega
- Plateforme Vecteurs Viraux et Transfert de Gènes, SFR Necker, US 24, UMS 3633, 75014 Paris, France
| | - Valeria Morante
- Department of Infection Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Donatella Negri
- Department of Infection Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Cara
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Gilles Dadaglio
- Institut Pasteur, Unité de Régulation Immunitaire et Vaccinologie, Equipe Labellisée Ligue Contre le Cancer, 75015 Paris, France; INSERM U1041, 75015 Paris, France.
| | - Claude Leclerc
- Institut Pasteur, Unité de Régulation Immunitaire et Vaccinologie, Equipe Labellisée Ligue Contre le Cancer, 75015 Paris, France; INSERM U1041, 75015 Paris, France.
| |
Collapse
|
133
|
Abstract
Immunity to malaria has been linked to the availability and function of helper CD4+ T cells, cytotoxic CD8+ T cells and γδ T cells that can respond to both the asymptomatic liver stage and the symptomatic blood stage of Plasmodium sp. infection. These T cell responses are also thought to be modulated by regulatory T cells. However, the precise mechanisms governing the development and function of Plasmodium-specific T cells and their capacity to form tissue-resident and long-lived memory populations are less well understood. The field has arrived at a point where the push for vaccines that exploit T cell-mediated immunity to malaria has made it imperative to define and reconcile the mechanisms that regulate the development and functions of Plasmodium-specific T cells. Here, we review our current understanding of the mechanisms by which T cell subsets orchestrate host resistance to Plasmodium infection on the basis of observational and mechanistic studies in humans, non-human primates and rodent models. We also examine the potential of new experimental strategies and human infection systems to inform a new generation of approaches to harness T cell responses against malaria.
Collapse
|
134
|
Ueki H, Wang IH, Zhao D, Gunzer M, Kawaoka Y. Multicolor two-photon imaging of in vivo cellular pathophysiology upon influenza virus infection using the two-photon IMPRESS. Nat Protoc 2020; 15:1041-1065. [PMID: 31996843 PMCID: PMC7086515 DOI: 10.1038/s41596-019-0275-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/03/2019] [Indexed: 12/14/2022]
Abstract
In vivo two-photon imaging is a valuable technique for studies of viral pathogenesis and host responses to infection in vivo. In this protocol, we describe a methodology for analyzing influenza virus-infected lung in vivo by two-photon imaging microscopy. We describe the surgical procedure, how to stabilize the lung, and an approach to analyzing the data. Further, we provide a database of fluorescent dyes, antibodies, and reporter mouse lines that can be used in combination with a reporter influenza virus (Color-flu) for multicolor analysis. Setup of this model typically takes ~30 min and enables the observation of influenza virus-infected lungs for >4 h during the acute phase of the inflammation and at least 1 h in the lethal phase. This imaging system, which we termed two-photon IMPRESS (imaging pathophysiology research system), is broadly applicable to analyses of other respiratory pathogens and reveals disease progression at the cellular level in vivo.
Collapse
Affiliation(s)
- Hiroshi Ueki
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - I-Hsuan Wang
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Dongming Zhao
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Yoshihiro Kawaoka
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
- Department of Special Pathogens, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| |
Collapse
|
135
|
Audsley KM, McDonnell AM, Waithman J. Cross-Presenting XCR1 + Dendritic Cells as Targets for Cancer Immunotherapy. Cells 2020; 9:cells9030565. [PMID: 32121071 PMCID: PMC7140519 DOI: 10.3390/cells9030565] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/14/2020] [Accepted: 02/25/2020] [Indexed: 12/11/2022] Open
Abstract
The use of dendritic cells (DCs) to generate effective anti-tumor T cell immunity has garnered much attention over the last thirty-plus years. Despite this, limited clinical benefit has been demonstrated thus far. There has been a revival of interest in DC-based treatment strategies following the remarkable patient responses observed with novel checkpoint blockade therapies, due to the potential for synergistic treatment. Cross-presenting DCs are recognized for their ability to prime CD8+ T cell responses to directly induce tumor death. Consequently, they are an attractive target for next-generation DC-based strategies. In this review, we define the universal classification system for cross-presenting DCs, and the vital role of this subset in mediating anti-tumor immunity. Furthermore, we will detail methods of targeting these DCs both ex vivo and in vivo to boost their function and drive effective anti-tumor responses.
Collapse
Affiliation(s)
- Katherine M. Audsley
- Telethon Kids Institute, University of Western Australia, Perth Children’s Hospital, Nedlands, WA 6009, Australia
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA 6009, Australia
- Correspondence: (K.M.A.); (A.M.M.); (J.W.); Tel.: +61-08-6319-1198 (K.M.A); +61-08-6319-1744 (J.W.)
| | - Alison M. McDonnell
- Telethon Kids Institute, University of Western Australia, Perth Children’s Hospital, Nedlands, WA 6009, Australia
- National Centre for Asbestos Related Diseases, The University of Western Australia, QEII Medical Centre, Nedlands, WA 6009, Australia
- Correspondence: (K.M.A.); (A.M.M.); (J.W.); Tel.: +61-08-6319-1198 (K.M.A); +61-08-6319-1744 (J.W.)
| | - Jason Waithman
- Telethon Kids Institute, University of Western Australia, Perth Children’s Hospital, Nedlands, WA 6009, Australia
- Correspondence: (K.M.A.); (A.M.M.); (J.W.); Tel.: +61-08-6319-1198 (K.M.A); +61-08-6319-1744 (J.W.)
| |
Collapse
|
136
|
Monti M, Consoli F, Vescovi R, Bugatti M, Vermi W. Human Plasmacytoid Dendritic Cells and Cutaneous Melanoma. Cells 2020; 9:E417. [PMID: 32054102 PMCID: PMC7072514 DOI: 10.3390/cells9020417] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/05/2020] [Accepted: 02/07/2020] [Indexed: 12/12/2022] Open
Abstract
The prognosis of metastatic melanoma (MM) patients has remained poor for a long time. However, the recent introduction of effective target therapies (BRAF and MEK inhibitors for BRAFV600-mutated MM) and immunotherapies (anti-CTLA-4 and anti-PD-1) has significantly improved the survival of MM patients. Notably, all these responses are highly dependent on the fitness of the host immune system, including the innate compartment. Among immune cells involved in cancer immunity, properly activated plasmacytoid dendritic cells (pDCs) exert an important role, bridging the innate and adaptive immune responses and directly eliminating cancer cells. A distinctive feature of pDCs is the production of high amount of type I Interferon (I-IFN), through the Toll-like receptor (TLR) 7 and 9 signaling pathway activation. However, published data indicate that melanoma-associated escape mechanisms are in place to hijack pDC functions. We have recently reported that pDC recruitment is recurrent in the early phases of melanoma, but the entire pDC compartment collapses over melanoma progression. Here, we summarize recent advances on pDC biology and function within the context of melanoma immunity.
Collapse
Affiliation(s)
- Matilde Monti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (M.B.)
| | - Francesca Consoli
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, Medical Oncology, University of Brescia at ASST-Spedali Civili, 25123 Brescia, Italy;
| | - Raffaella Vescovi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (M.B.)
| | - Mattia Bugatti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (M.B.)
| | - William Vermi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (M.B.)
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
137
|
Prokhnevska N, Emerson DA, Kissick HT, Redmond WL. Immunological Complexity of the Prostate Cancer Microenvironment Influences the Response to Immunotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1210:121-147. [PMID: 31900908 DOI: 10.1007/978-3-030-32656-2_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Prostate cancer is one of the most common cancers in men and a leading cause of cancer-related death. Recent advances in the treatment of advanced prostate cancer, including the use of more potent and selective inhibitors of the androgen signaling pathway, have provided significant clinical benefit for men with metastatic castration-resistant prostate cancer (mCRPC). However, most patients develop progressive lethal disease, highlighting the need for more effective treatments. One such approach is immunotherapy, which harness the power of the patient's immune system to identify and destroy cancer cells through the activation of cytotoxic CD8 T cells specific for tumor antigens. Although immunotherapy, particularly checkpoint blockade, can induce significant clinical responses in patients with solid tumors or hematological malignancies, minimal efficacy has been observed in men with mCRPC. In the current review, we discuss our current understanding of the immunological complexity of the immunosuppressive prostate cancer microenvironment, preclinical models of prostate cancer, and recent advances in immunotherapy clinical trials to improve outcomes for men with mCRPC.
Collapse
Affiliation(s)
| | - Dana A Emerson
- Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA.,Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | | | - William L Redmond
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA.
| |
Collapse
|
138
|
Li F, Huang Q, Luster TA, Hu H, Zhang H, Ng WL, Khodadadi-Jamayran A, Wang W, Chen T, Deng J, Ranieri M, Fang Z, Pyon V, Dowling CM, Bagdatlioglu E, Almonte C, Labbe K, Silver H, Rabin AR, Jani K, Tsirigos A, Papagiannakopoulos T, Hammerman PS, Velcheti V, Freeman GJ, Qi J, Miller G, Wong KK. In Vivo Epigenetic CRISPR Screen Identifies Asf1a as an Immunotherapeutic Target in Kras-Mutant Lung Adenocarcinoma. Cancer Discov 2020; 10:270-287. [PMID: 31744829 PMCID: PMC7007372 DOI: 10.1158/2159-8290.cd-19-0780] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/11/2019] [Accepted: 11/13/2019] [Indexed: 11/16/2022]
Abstract
Despite substantial progress in lung cancer immunotherapy, the overall response rate in patients with KRAS-mutant lung adenocarcinoma (LUAD) remains low. Combining standard immunotherapy with adjuvant approaches that enhance adaptive immune responses-such as epigenetic modulation of antitumor immunity-is therefore an attractive strategy. To identify epigenetic regulators of tumor immunity, we constructed an epigenetic-focused single guide RNA library and performed an in vivo CRISPR screen in a Kras G12D/Trp53 -/- LUAD model. Our data showed that loss of the histone chaperone Asf1a in tumor cells sensitizes tumors to anti-PD-1 treatment. Mechanistic studies revealed that tumor cell-intrinsic Asf1a deficiency induced immunogenic macrophage differentiation in the tumor microenvironment by upregulating GM-CSF expression and potentiated T-cell activation in combination with anti-PD-1. Our results provide a rationale for a novel combination therapy consisting of ASF1A inhibition and anti-PD-1 immunotherapy. SIGNIFICANCE: Using an in vivo epigenetic CRISPR screen, we identified Asf1a as a critical regulator of LUAD sensitivity to anti-PD-1 therapy. Asf1a deficiency synergized with anti-PD-1 immunotherapy by promoting M1-like macrophage polarization and T-cell activation. Thus, we provide a new immunotherapeutic strategy for this subtype of patients with LUAD.See related commentary by Menzel and Black, p. 179.This article is highlighted in the In This Issue feature, p. 161.
Collapse
Affiliation(s)
- Fei Li
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Qingyuan Huang
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Troy A Luster
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Hai Hu
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Hua Zhang
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Wai-Lung Ng
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR
| | - Alireza Khodadadi-Jamayran
- Applied Bioinformatics Laboratories and Genome Technology Center, Division of Advanced Research Technologies, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Wei Wang
- S. Arthur Localio Laboratory, Department of Surgery, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Ting Chen
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Jiehui Deng
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Michela Ranieri
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Zhaoyuan Fang
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Val Pyon
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Catríona M Dowling
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Ece Bagdatlioglu
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Christina Almonte
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Kristen Labbe
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Heather Silver
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Alexandra R Rabin
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Kandarp Jani
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Aristotelis Tsirigos
- Applied Bioinformatics Laboratories and Genome Technology Center, Division of Advanced Research Technologies, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
- Department of Pathology, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Thales Papagiannakopoulos
- Department of Pathology, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Peter S Hammerman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Vamsidhar Velcheti
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jun Qi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - George Miller
- S. Arthur Localio Laboratory, Department of Surgery, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York.
| |
Collapse
|
139
|
Li Y, Zhang M, Wang X, Liu W, Wang H, Yang YG. Vaccination with CD47 deficient tumor cells elicits an antitumor immune response in mice. Nat Commun 2020; 11:581. [PMID: 31996683 PMCID: PMC6989506 DOI: 10.1038/s41467-019-14102-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 12/10/2019] [Indexed: 01/28/2023] Open
Abstract
Cancer cells are poorly immunogenic and have a wide range of mutations, which makes them unsuitable for use in vaccination treatment. Here, we show that elimination of CD47, a ligand for the myeloid cell inhibitory receptor SIRPα, from tumor cells by genetic deletion or antibody blocking, significantly improves the effectiveness of the immune response to tumour cells. In both solid and hematopoietic mouse tumor models, vaccination with tumor cells or tumor antigen-expressing cells, that lack CD47 or were pre-coated with anti-CD47 antibodies, achieved an antitumor immune response. The efficacy of this approach was synergistically enhanced when used in combination with anti-PD-1 antibodies. The induction of antitumor responses depends on SIRPα+CD11c+ DCs, which exhibit rapid expansion following introduction of CD47-deficient tumor cells. Our results indicate that CD47-deficient whole tumor cells can induce antitumor responses.
Collapse
Affiliation(s)
- Yang Li
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital, and Institute of Immunology, Jilin University, Changchun, China.,Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - Mingyou Zhang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital, and Institute of Immunology, Jilin University, Changchun, China.,Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - Xiaodan Wang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital, and Institute of Immunology, Jilin University, Changchun, China.,National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Wentao Liu
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital, and Institute of Immunology, Jilin University, Changchun, China.,National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Hui Wang
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital, and Institute of Immunology, Jilin University, Changchun, China. .,Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA. .,National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China. .,International Center of Future Science, Jilin University, Changchun, China.
| |
Collapse
|
140
|
Alam MS, Cavanaugh C, Pereira M, Babu U, Williams K. Susceptibility of aging mice to listeriosis: Role of anti-inflammatory responses with enhanced Treg-cell expression of CD39/CD73 and Th-17 cells. Int J Med Microbiol 2020; 310:151397. [PMID: 31974050 DOI: 10.1016/j.ijmm.2020.151397] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 11/14/2019] [Accepted: 12/18/2019] [Indexed: 12/22/2022] Open
Abstract
Foodborne Listeria monocytogenes (Lm) causes serious illness and death in immunosuppressed hosts, including the elderly population. We investigated Lm susceptibility and inflammatory cytokines in geriatric mice. Young-adult and old mice were gavaged with a Lm strain Lmo-InlAm. Tissues were assayed for Lm burden and splenocytes were analyzed for Th1/Th2/Th17/Treg responses and expression of CD39 and CD73. Old Lm-infected mice lost body-weight dose-dependently, had higher Lm colonization, and showed higher inflammatory responses than Lm-infected young-adult mice. After infection, IL-17 levels increased significantly in old mice whereas IFN-γ levels were unchanged. Levels of IL-10 and Treg cells were increased in infected old mice as compared to infected young-adult mice. Age-dependent enhanced expression of CD39/CD73 was observed in purified Treg prior to infection, suggesting increased baseline adenosine production in old mice. Lm lysate-treated splenocytes from older mice produced significantly higher levels of IL-10, IL17, and IL-1β, produced less IFN-γ and IL-2, and proliferated less than splenocytes from young-adult mice. Data suggests that older mice maybe more susceptible to Lm infection due to an imbalance of Th cell responses with disproportionate and persistent anti-inflammatory responses. Lm infection enhanced differentiation of proinflammatory Th17 cells, which may also exacerbate pathological responses during listeriosis.
Collapse
Affiliation(s)
- M Samiul Alam
- Immunobiology Branch, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, Laurel, MD, 20708, USA.
| | - Christopher Cavanaugh
- Immunobiology Branch, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, Laurel, MD, 20708, USA
| | - Marion Pereira
- Immunobiology Branch, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, Laurel, MD, 20708, USA
| | - Uma Babu
- Immunobiology Branch, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, Laurel, MD, 20708, USA
| | - Kristina Williams
- Immunobiology Branch, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, Laurel, MD, 20708, USA
| |
Collapse
|
141
|
Cruz FM, Colbert JD, Rock KL. The GTPase Rab39a promotes phagosome maturation into MHC-I antigen-presenting compartments. EMBO J 2020; 39:e102020. [PMID: 31821587 PMCID: PMC6960445 DOI: 10.15252/embj.2019102020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 11/04/2019] [Accepted: 11/12/2019] [Indexed: 12/21/2022] Open
Abstract
For CD8 T lymphocytes to mount responses to cancer and virally-infected cells, dendritic cells must capture antigens present in tissues and display them as peptides bound to MHC-I molecules. This is most often accomplished through a pathway called antigen cross-presentation (XPT). Here, we report that the vesicular trafficking protein Rab39a is needed for optimal cross-presentation by dendritic cells in vitro and cross-priming of CD8 T cells in vivo. Without Rab39a, MHC-I presentation of intraphagosomal peptides is inhibited, indicating that Rab39a converts phagosomes into peptide-loading compartments. In this process, Rab39a promotes the delivery of MHC-I molecules from the endoplasmic reticulum (ER) to phagosomes, and increases the levels of peptide-empty MHC-I conformers that can be loaded with peptide in this compartment. Rab39a also increases the levels of Sec22b and NOX2, previously recognized to participate in cross-presentation, on phagosomes, thereby filling in a missing link into how phagosomes mature into cross-presenting vesicles.
Collapse
Affiliation(s)
- Freidrich M Cruz
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Jeff D Colbert
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Kenneth L Rock
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| |
Collapse
|
142
|
Gross-Vered M, Trzebanski S, Shemer A, Bernshtein B, Curato C, Stelzer G, Salame TM, David E, Boura-Halfon S, Chappell-Maor L, Leshkowitz D, Jung S. Defining murine monocyte differentiation into colonic and ileal macrophages. eLife 2020; 9:e49998. [PMID: 31916932 PMCID: PMC6952180 DOI: 10.7554/elife.49998] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 11/27/2019] [Indexed: 12/22/2022] Open
Abstract
Monocytes are circulating short-lived macrophage precursors that are recruited on demand from the blood to sites of inflammation and challenge. In steady state, classical monocytes give rise to vasculature-resident cells that patrol the luminal side of the endothelium. In addition, classical monocytes feed macrophage compartments of selected organs, including barrier tissues, such as the skin and intestine, as well as the heart. Monocyte differentiation under conditions of inflammation has been studied in considerable detail. In contrast, monocyte differentiation under non-inflammatory conditions remains less well understood. Here we took advantage of a combination of cell ablation and precursor engraftment to investigate the generation of gut macrophages from monocytes. Collectively, we identify factors associated with the gradual adaptation of monocytes to tissue residency. Moreover, comparison of monocyte differentiation into the colon and ileum-resident macrophages revealed the graduated acquisition of gut segment-specific gene expression signatures.
Collapse
Affiliation(s)
- Mor Gross-Vered
- Department of ImmunologyWeizmann Institute of ScienceRehovotIsrael
| | | | - Anat Shemer
- Department of ImmunologyWeizmann Institute of ScienceRehovotIsrael
| | - Biana Bernshtein
- Department of ImmunologyWeizmann Institute of ScienceRehovotIsrael
| | - Caterina Curato
- Department of ImmunologyWeizmann Institute of ScienceRehovotIsrael
| | - Gil Stelzer
- Bioinformatics Unit, Life Science Core FacilitiesWeizmann Institute of ScienceRehovotIsrael
| | - Tomer-Meir Salame
- Bioinformatics Unit, Life Science Core FacilitiesWeizmann Institute of ScienceRehovotIsrael
| | - Eyal David
- Department of ImmunologyWeizmann Institute of ScienceRehovotIsrael
| | | | | | - Dena Leshkowitz
- Bioinformatics Unit, Life Science Core FacilitiesWeizmann Institute of ScienceRehovotIsrael
| | - Steffen Jung
- Department of ImmunologyWeizmann Institute of ScienceRehovotIsrael
| |
Collapse
|
143
|
Kretschmer L, Flossdorf M, Mir J, Cho YL, Plambeck M, Treise I, Toska A, Heinzel S, Schiemann M, Busch DH, Buchholz VR. Differential expansion of T central memory precursor and effector subsets is regulated by division speed. Nat Commun 2020; 11:113. [PMID: 31913278 PMCID: PMC6949285 DOI: 10.1038/s41467-019-13788-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 11/26/2019] [Indexed: 12/14/2022] Open
Abstract
While antigen-primed T cells proliferate at speeds close to the physiologic maximum of mammalian cells, T cell memory is maintained in the absence of antigen by rare cell divisions. The transition between these distinct proliferative programs has been difficult to resolve via population-based analyses. Here, we computationally reconstruct the proliferative history of single CD8+ T cells upon vaccination and measure the division speed of emerging T cell subsets in vivo. We find that slower cycling central memory precursors, characterized by an elongated G1 phase, segregate early from the bulk of rapidly dividing effector subsets, and further slow-down their cell cycle upon premature removal of antigenic stimuli. In contrast, curtailed availability of inflammatory stimuli selectively restrains effector T cell proliferation due to reduced receptivity for interleukin-2. In line with these findings, persistence of antigenic but not inflammatory stimuli throughout clonal expansion critically determines the later size of the memory compartment.
Collapse
Affiliation(s)
- Lorenz Kretschmer
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Michael Flossdorf
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Jonas Mir
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Yi-Li Cho
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Marten Plambeck
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Irina Treise
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany.,German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Albulena Toska
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Susanne Heinzel
- Division of Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Matthias Schiemann
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany. .,German Center for Infection Research (DZIF), Partner site Munich, Munich, Germany.
| | - Veit R Buchholz
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany.
| |
Collapse
|
144
|
Miki H, Tahara-Hanaoka S, Almeida MS, Hitomi K, Shibagaki S, Kanemaru K, Lin YH, Iwata K, Miyake S, Shibayama S, Sumida T, Shibuya K, Shibuya A. Allergin-1 Immunoreceptor Suppresses House Dust Mite-Induced Allergic Airway Inflammation. THE JOURNAL OF IMMUNOLOGY 2020; 204:753-762. [PMID: 31900344 DOI: 10.4049/jimmunol.1900180] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 12/02/2019] [Indexed: 01/04/2023]
Abstract
House dust mite (HDM) allergens are leading causes of allergic asthma characterized by Th2 responses. The lung-resident CD11b+ dendritic cells (DCs) play a key role in Th2 cell development in HDM-induced allergic asthma. However, the regulatory mechanism of HDM-induced CD11b+ DC activation remains incompletely understood. In this study, we demonstrate that mice deficient in an inhibitory immunoreceptor, Allergin-1, showed exacerbated HDM-induced airway eosinophilia and serum IgE elevation. By using bone marrow-chimeric mice that were sensitized with adoptively transferred HDM-stimulated wild-type or Allergin-1-deficient CD11b+ bone marrow-derived cultured DCs (BMDCs), followed by challenge with HDM, we show that Allergin-1 on the BMDCs suppressed HDM-induced allergic airway inflammation. We also show that Allergin-1 suppressed HDM-induced PGE2 production from CD11b+ BMDCs by inhibiting Syk tyrosine kinase activation through recruitment of SHP-1, subsequently leading to negative regulation of Th2 responses. These results suggest that Allergin-1 plays an important role in regulation of HDM-induced allergic airway inflammation.
Collapse
Affiliation(s)
- Haruka Miki
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan.,Department of Internal Medicine, Faculty of Medicine, Tsukuba Advanced Research Alliance, R&D Center for Innovative Drug Discovery, Tsukuba, Ibaraki 305-8575, Japan
| | - Satoko Tahara-Hanaoka
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; .,Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Mariana Silva Almeida
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Kaori Hitomi
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Shohei Shibagaki
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Kazumasa Kanemaru
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yu-Hsien Lin
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan.,Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; and
| | - Kanako Iwata
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Shota Miyake
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Shiro Shibayama
- Research Center of Immunology, Tsukuba Institute, Ono Pharmaceutical Co., Ltd., Tsukuba, Ibaraki 300-4247, Japan
| | - Takayuki Sumida
- Department of Internal Medicine, Faculty of Medicine, Tsukuba Advanced Research Alliance, R&D Center for Innovative Drug Discovery, Tsukuba, Ibaraki 305-8575, Japan
| | - Kazuko Shibuya
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan.,Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Akira Shibuya
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; .,Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
145
|
Benites BD, Alvarez MC, Saad STO. Small Particles, Big Effects: The Interplay Between Exosomes and Dendritic Cells in Antitumor Immunity and Immunotherapy. Cells 2019; 8:E1648. [PMID: 31888159 PMCID: PMC6952774 DOI: 10.3390/cells8121648] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/07/2019] [Accepted: 12/10/2019] [Indexed: 12/18/2022] Open
Abstract
Dendritic cells play a fundamental role in the antitumor immunity cycle, and the loss of their antigen-presenting function is a recognized mechanism of tumor evasion. We have recently demonstrated the effect of exosomes extracted from serum of patients with acute myeloid leukemia as important inducers of dendritic cell immunotolerance, and several other works have recently demonstrated the effects of these nanoparticles on immunity to other tumor types as well. The aim of this review was to highlight the recent findings on the effects of tumor exosomes on dendritic cell functions, the mechanisms by which they can lead to tumor evasion, and their manipulation as a possible strategy in cancer treatment.
Collapse
Affiliation(s)
- Bruno Deltreggia Benites
- Hematology and Transfusion Medicine Center, University of Campinas, Campinas 13083-970, Brazil; (M.C.A.); (S.T.O.S.)
| | | | | |
Collapse
|
146
|
Sil P, Suwanpradid J, Muse G, Gruzdev A, Liu L, Corcoran DL, Willson CJ, Janardhan K, Grimm S, Myers P, Degraff LM, MacLeod AS, Martinez J. Noncanonical autophagy in dermal dendritic cells mediates immunosuppressive effects of UV exposure. J Allergy Clin Immunol 2019; 145:1389-1405. [PMID: 31837371 DOI: 10.1016/j.jaci.2019.11.041] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/10/2019] [Accepted: 11/05/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Control of the inflammatory response is critical to maintaining homeostasis, and failure to do so contributes to the burden of chronic inflammation associated with several disease states. The mechanisms that underlie immunosuppression, however, remain largely unknown. Although defects in autophagy machinery have been associated with inflammatory pathologic conditions, we now appreciate that autophagic components participate in noncanonical pathways distinct from classical autophagy. We have previously demonstrated that LC3-associated phagocytosis (LAP), a noncanonical autophagic process dependent on Rubicon (rubicon autophagy regulator [RUBCN]), contributes to immunosuppression. OBJECTIVE We used Rubcn-/- mice to examine the role of the LAP pathway in mediating the UV-induced immunotolerant program in a model of contact hypersensitivity (CHS). METHODS Flow cytometry and transcriptional analysis were used to measure immune cell infiltration and activation in the skin of Rubcn+/+ and Rubcn-/- mice during the CHS response. RESULTS Here, we demonstrate that LAP is required for UV-induced immunosuppression and that UV exposure induces a broadly anti-inflammatory transcriptional program dependent on Rubicon. Rubcn-/- mice are resistant to UV-induced immunosuppression and instead display exaggerated inflammation in a model of CHS. Specifically, RUBCN deficiency in CD301b+ dermal dendritic cells results in their increased antigen presentation capacity and subsequent hyperactivation of the CD8+ T-cell response. CONCLUSIONS LAP functions to limit the immune response and is critical in maintaining the balance between homeostasis and inflammation.
Collapse
Affiliation(s)
- Payel Sil
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | | | - Ginger Muse
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Artiom Gruzdev
- Knockout Mouse Core Laboratory, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Liwen Liu
- Molecular Genomics Core Laboratory, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - David L Corcoran
- Duke Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC
| | | | | | - Sara Grimm
- Division of Intramural Research, Research Triangle Park, NC
| | - Page Myers
- Comparative Medicine Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Laura Miller Degraff
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Amanda S MacLeod
- Department of Dermatology, Duke University, Durham, NC; Department of Immunology, Duke University, Durham, NC; Department of Molecular Genetics and Microbiology, Duke University, Durham, NC
| | - Jennifer Martinez
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC.
| |
Collapse
|
147
|
Zhao H, Tian X, He L, Li Y, Pu W, Liu Q, Tang J, Wu J, Cheng X, Liu Y, Zhou Q, Tan Z, Bai F, Xu F, Smart N, Zhou B. Apj + Vessels Drive Tumor Growth and Represent a Tractable Therapeutic Target. Cell Rep 2019; 25:1241-1254.e5. [PMID: 30380415 DOI: 10.1016/j.celrep.2018.10.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 08/20/2018] [Accepted: 10/03/2018] [Indexed: 02/02/2023] Open
Abstract
Identification of cellular surface markers that distinguish tumorous from normal vasculature is important for the development of tumor vessel-targeted therapy. Here, we show that Apj, a G protein-coupled receptor, is highly enriched in tumor endothelial cells but absent from most endothelial cells of adult tissues in homeostasis. By genetic targeting using Apj-CreER and Apj-DTRGFP-Luciferase, we demonstrated that hypoxia-VEGF signaling drives expansion of Apj+ tumor vessels and that targeting of these vessels, genetically and pharmacologically, remarkably inhibits tumor angiogenesis and restricts tumor growth. These in vivo findings implicate Apj+ vessels as a key driver of pathological angiogenesis and identify Apj+ endothelial cells as an important therapeutic target for the anti-angiogenic treatment of tumors.
Collapse
Affiliation(s)
- Huan Zhao
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China; Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xueying Tian
- Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, China
| | - Lingjuan He
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China; Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yan Li
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China; Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wenjuan Pu
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China; Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Qiaozhen Liu
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China; Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Juan Tang
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China; Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jiaying Wu
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xin Cheng
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yang Liu
- Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, Peking University, Beijing, 100871, China
| | - Qingtong Zhou
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, China
| | - Zhen Tan
- Department of Pediatric Hematology/Oncology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, China
| | - Fan Bai
- Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, Peking University, Beijing, 100871, China
| | - Fei Xu
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Nicola Smart
- British Heart Foundation Centre of Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Bin Zhou
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China; Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China; Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
148
|
Barak AF, Lewinsky H, Perpinial M, Huber V, Radomir L, Kramer MP, Sever L, Wolf Y, Shapiro M, Herishanu Y, Jung S, Becker-Herman S, Shachar I. Bone marrow dendritic cells support the survival of chronic lymphocytic leukemia cells in a CD84 dependent manner. Oncogene 2019; 39:1997-2008. [PMID: 31772329 DOI: 10.1038/s41388-019-1121-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 11/11/2019] [Accepted: 11/13/2019] [Indexed: 11/09/2022]
Abstract
Chronic lymphocytic leukemia (CLL) is a malignancy of mature B lymphocytes. The microenvironment of the CLL cells is a vital element in the regulation of the survival of these malignant cells. CLL cell longevity is dependent on external signals, originating from cells in their microenvironment including secreted and surface-bound factors. Dendritic cells (DCs) play an important part in tumor microenvironment, but their role in the CLL bone marrow (BM) niche has not been studied. We show here that CLL cells induce accumulation of bone marrow dendritic cells (BMDCs). Depletion of this population attenuates disease expansion. Our results show that the support of the microenvironment is partly dependent on CD84, a cell surface molecule belonging to the Signaling Lymphocyte Activating Molecule (SLAM) family of immunoreceptors. Our results suggest a novel therapeutic strategy whereby eliminating BMDCs or blocking the CD84 expressed on these cells may reduce the tumor load.
Collapse
Affiliation(s)
- Avital F Barak
- Department of Immunology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Hadas Lewinsky
- Department of Immunology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Michal Perpinial
- Department of Immunology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Victoria Huber
- Department of Immunology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Lihi Radomir
- Department of Immunology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Mattias P Kramer
- Department of Immunology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Lital Sever
- Department of Immunology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Yochai Wolf
- Department of Immunology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Mika Shapiro
- Department of Hematology, Tel-Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Yair Herishanu
- Department of Hematology, Tel-Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | | | - Idit Shachar
- Department of Immunology, Weizmann Institute of Science, Rehovot, 76100, Israel.
| |
Collapse
|
149
|
Cross-presentation of Exogenous Antigens. Transfus Clin Biol 2019; 26:346-351. [DOI: 10.1016/j.tracli.2019.01.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 01/25/2019] [Indexed: 01/07/2023]
|
150
|
Kumar R, Loughland JR, Ng SS, Boyle MJ, Engwerda CR. The regulation of CD4
+
T cells during malaria. Immunol Rev 2019; 293:70-87. [DOI: 10.1111/imr.12804] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 09/04/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Rajiv Kumar
- Centre of Experimental Medicine and Surgery Institute of Medical Sciences Banaras Hindu University Varanasi UP India
- Department of Medicine Institute of Medical Sciences Banaras Hindu University Varanasi UP India
| | - Jessica R. Loughland
- Human Malaria Immunology Laboratory QIMR Berghofer Medical Research Institute Brisbane Australia
| | - Susanna S. Ng
- Immunology and Infection Laboratory QIMR Berghofer Medical Research Institute Brisbane Australia
| | - Michelle J. Boyle
- Human Malaria Immunology Laboratory QIMR Berghofer Medical Research Institute Brisbane Australia
| | - Christian R. Engwerda
- Immunology and Infection Laboratory QIMR Berghofer Medical Research Institute Brisbane Australia
| |
Collapse
|