1
|
Lawrence LA, Williams ME, Vidal P, Varughese RS, Li ZRT, Chen TD, Roy MA, Tuske SC, Lowen AC, Scharer CD, Shafer WM, Swaims-Kohlmeier A. Murine modeling of menstruation identifies immune correlates of protection during Chlamydia muridarum challenge. PLoS Pathog 2025; 21:e1012276. [PMID: 40478914 DOI: 10.1371/journal.ppat.1012276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 05/06/2025] [Indexed: 06/11/2025] Open
Abstract
The menstrual cycle influences the risk of acquiring sexually transmitted infections (STIs), including those caused by the pathogen Chlamydia trachomatis (C. trachomatis). However, the underlying immune contributions are poorly defined. A mouse model simulating the repetitive immune-mediated process of menstruation could provide valuable insights into tissue-specific determinants of protection against chlamydial infection within the cervicovaginal and uterine mucosae of the female reproductive tract (FRT). Here, we used the pseudopregnancy approach for inducing menstruation in naïve C57Bl/6 mice and performed vaginal challenge with Chlamydia muridarum (C. muridarum) over the course of decidualization, endometrial tissue remodeling, and menstruation. This strategy identified that a time point over pseudopregnancy corresponding to the late luteal phase of the menstrual cycle correlated with reduced bacterial burden. By evaluating the early infection site following challenge at this time point, we found that a greater abundance of NK cell populations and proinflammatory signaling, including IFNγ, were strongly correlated with protection. FRT immune profiling in uninfected mice over pseudopregnancy or in pig-tailed macaques over the menstrual cycle identified periodic NK cell infiltration into the cervicovaginal tissues and luminal surface occurring over a similar time frame. Notably, these cell populations were transcriptionally distinct and enriched for programs associated with NK cell effector functions. Depletion of FRT NK cells during the late luteal phase resulted in a loss of protection, enabling productive infection following C. muridarum challenge. This study shows that the pseudopregnancy murine menstruation model recapitulates dynamic changes occurring in mucosal immune states throughout the FRT as a result of endometrial remodeling and identifies NK cell localization at the FRT barrier site of pathogen exposure as essential for immune protection against primary C. muridarum infection.
Collapse
Affiliation(s)
- Laurel A Lawrence
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - M Elliott Williams
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Paola Vidal
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Richa S Varughese
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Zheng-Rong Tiger Li
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Thien Duy Chen
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Melissa A Roy
- Division of Pathology Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Steven C Tuske
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Anice C Lowen
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - William M Shafer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Laboratories of Bacterial Pathogenesis, Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States of America
| | - Alison Swaims-Kohlmeier
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of GYNOB, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Division of HIV Prevention Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| |
Collapse
|
2
|
He D, Dai W, Cui Y, Gao R, Yang X, Li W, Zhou J, Yin S, Kuang T, Zhu J, Luo L, Wang R, Qu Y, Yun C, Liu Z, Diao X, Ma X, Liang H, Wang F. Diamine oxidase acts as a novel risk factor in abnormal inflammation via mediating "cytosolic ROS-autophagy-IFN-γ" axis in NK cells. Life Sci 2025; 377:123775. [PMID: 40449879 DOI: 10.1016/j.lfs.2025.123775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 05/21/2025] [Accepted: 05/27/2025] [Indexed: 06/03/2025]
Abstract
AIMS Diamine oxidase (DAO), a well-established biomarker for intestinal damage, histamine intolerance or tumorigenesis, has rarely been reported in immune regulation. This study aimed to identify DAO as a critical enhancer of abnormal inflammation by promoting interferon-gamma (IFN-γ) production from natural killer (NK) cells. MAIN METHODS Clinical bioinformatics analyzed aoc1 (DAO-coding gene) expression in PBMCs from patients with inflammatory diseases. Murine models (LPS-induced systemic inflammation, sepsis, DSS-induced colitis) using DAO-/- mice, alongside DAO-/- NK92 cells and DAO inhibitor DIZE, were employed for phenotypic validation. Cellular profiling, bone marrow chimeras, reciprocal transplantation, RNA-sequence, non-targeted metabolomics, and flow cytometry were utilized to dissect DAO's mechanisms in NK cells. KEY FINDINGS DAO deficiency protected mice from inflammatory pathology by suppressing IFN-γ production. NK cells were identified as the primary target cells during the process, with DAO acting intracellularly to promote IFN-γ via a reactive oxygen species (ROS)-autophagy axis. DAO-derived ROS, distinct from mitochondrial or NOX2 sources, enhanced autophagic flux during NK activation, enabling IFN-γ biosynthesis. DAO did not affect NK homeostasis, including maturation, proliferation, or receptor expressions. SIGNIFICANCE DAO is a novel risk factor in inflammatory diseases, driving IFN-γ production through ROS-autophagy signaling in NK cells. Targeting DAO may offer therapeutic strategies for conditions involving dysregulated IFN-γ responses, including sepsis, colitis, and autoimmune disorders.
Collapse
Affiliation(s)
- Dongmei He
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Weihong Dai
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China; Emergency of The Second Affiliated Hospital, Hainan Medical University, Haikou 571100, Hainan, China
| | - Yiqin Cui
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Rui Gao
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Xue Yang
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Wei Li
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Jing Zhou
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Shuangqin Yin
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Tianyin Kuang
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Junyu Zhu
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Li Luo
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Rixing Wang
- Emergency of The Second Affiliated Hospital, Hainan Medical University, Haikou 571100, Hainan, China
| | - Ye Qu
- Emergency of The Second Affiliated Hospital, Hainan Medical University, Haikou 571100, Hainan, China
| | - Caihong Yun
- Emergency of The Second Affiliated Hospital, Hainan Medical University, Haikou 571100, Hainan, China
| | - Zhuli Liu
- Emergency of The Second Affiliated Hospital, Hainan Medical University, Haikou 571100, Hainan, China
| | - Xiaoyan Diao
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Xiaoyuan Ma
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China.
| | - Huaping Liang
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China.
| | - Fangjie Wang
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China.
| |
Collapse
|
3
|
Lukas D, Liu X, Reibetanz M, Könen-Waisman S, Bopp T, Bohn T, Vivier E, Gasteiger G, Waisman A, von Stebut E. Innate lymphoid cell (ILC) 1- and NK cell-derived, early IFNγ release depends on ICER and promotes protection against Leishmania major infection. J Invest Dermatol 2025:S0022-202X(25)00477-4. [PMID: 40378947 DOI: 10.1016/j.jid.2025.04.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/06/2025] [Accepted: 04/08/2025] [Indexed: 05/19/2025]
Abstract
Innate lymphoid cells (ILCs) participate in different skin diseases. Cutaneous leishmaniasis evokes Th1/Tc1-dominated immunity, while in immune-compromised individuals, a Th2/Treg/Th17-immune response dominates. Only few prior studies investigated the role of ILC in leishmaniasis. We show that following physiologic low dose infection with Leishmania major, both lesional NK cell and ILC1 numbers strongly increase. In addition, early lesional IFNγ production derives from type I ILCs. Genetic ablation of both NK cells and ILC1 (NK/ILC1Δ mice) led to reduced early IFNγ expression with increased pathology, higher parasite burdens and delayed recovery. Furthermore, expression of inducible cAMP early repressor (ICER) is important for disease outcome, as ICER-/- mice exhibited significantly larger lesions. Interestingly, mice that lack ICER specifically in NK cells and ILC1 phenocopied the worsened disease outcome of ICER-/- mice, while ICER deficiency in T cells or macrophages alone failed to do so. In line, ICER deficiency in NK cells/ILC1 resulted in higher lesional parasite burden with fewer IFNγ-positive ILC1 compared to control animals. Thus, our data shows that both NK cells and ILC1 contribute to early parasite control by releasing IFNγ. ICER expression by ILC1 promotes recruitment of IFNγ+ ILC1 in Leishmania infections important for development of protection against this important pathogen.
Collapse
Affiliation(s)
- Dominika Lukas
- Department of Dermatology, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany
| | - Xinyuan Liu
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Marion Reibetanz
- Department of Dermatology, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany
| | - Stephanie Könen-Waisman
- Department of Dermatology, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Toszka Bohn
- Institute for Immunology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Eric Vivier
- Innate Pharma, Marseille, France; Aix-Marseille University, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France; APHM, Hôpital de la Timone, Marseille-Immunopôle, Marseille, France
| | - Georg Gasteiger
- Max Planck Research Group at the Julius-Maximilians-Universität Wurzburg, Wurzburg Institute of Systems Immunology, 97078 Wurzburg, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Esther von Stebut
- Department of Dermatology, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany.
| |
Collapse
|
4
|
Paton H, Sarkar P, Gurung P. An overview of host immune responses against Leishmania spp. infections. Hum Mol Genet 2025:ddaf043. [PMID: 40287829 DOI: 10.1093/hmg/ddaf043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Leishmania spp. infections pose a significant global health challenge, affecting approximately 1 billion people across more than 88 endemic countries. This unicellular, obligate intracellular parasite causes a spectrum of diseases, ranging from localized cutaneous lesions to systemic visceral infections. Despite advancements in modern medicine and increased understanding of the parasite's etiology and associated diseases, treatment options remain limited to pentavalent antimonials, liposomal amphotericin B, and miltefosine. A deeper understanding of the interactions between immune and non-immune cells involved in the clearance of Leishmania spp. infections could uncover novel therapeutic strategies for this debilitating disease. This review highlights recent progress in elucidating how various cell types contribute to the regulation and resolution of Leishmania spp. infections.
Collapse
Affiliation(s)
- Hanna Paton
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
- Immunology Graduate Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
| | - Prabuddha Sarkar
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
| | - Prajwal Gurung
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
- Immunology Graduate Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Center for Immunology and Immune Based Disease, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Iowa City Veterans Affairs (VA) Medical Center, 601 US-6, Iowa City, IA 52246, United States
| |
Collapse
|
5
|
Karati D, Meur S, Das S, Adak A, Mukherjee S. Peptide-based drugs in immunotherapy: current advances and future prospects. Med Oncol 2025; 42:177. [PMID: 40266466 DOI: 10.1007/s12032-025-02739-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/18/2025] [Indexed: 04/24/2025]
Abstract
In immunotherapy, peptide-based medications are showing great promise as a new class of therapies that can be used to treat autoimmune diseases, cancer, and other immune-related conditions. Peptides are being created for use in immunotherapy as vaccines, immunological modulators, and adjuvants because of their capacity to precisely alter immune responses. They can imitate endogenous signals or interact with immune cells, improving the body's capacity to identify and combat malignancies or reestablishing immunological tolerance in autoimmune disorders. Notably, peptide-based treatments have demonstrated promise in promoting tumor-specific immune responses and improving the effectiveness of already available immunotherapies, such as immune checkpoint inhibitors. Notwithstanding its potential, peptide-based medications' clinical translation is fraught with difficulties, such as those pertaining to immunogenicity, bioavailability, and peptide stability. Overcoming these obstacles has been made possible by developments in peptide engineering, including pharmacokinetic optimization, receptor-binding affinity enhancement, and the creation of innovative delivery systems. The targeted distribution and effectiveness of peptide medications can be improved by using liposomes, nanoparticles, and other delivery methods, increasing their therapeutic utility. With an emphasis on recent scientific developments, mechanisms of action, and therapeutic uses, this review examines the present status of peptide-based medications in immunotherapy. We also look at the obstacles that still need to be overcome in order to get peptide-based treatments from the lab to the clinic and offer suggestions for future research initiatives. By tackling these important problems, we hope to demonstrate how peptide-based medications have the ability to revolutionize immunotherapeutic treatment approaches.
Collapse
Affiliation(s)
- Dipanjan Karati
- Department of Pharmaceutical Technology, School of Pharmacy, Techno India University-TIU, Kolkata, West Bengal, 700091, India
| | - Shreyasi Meur
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata - Group of Institutions, Kolkata, West Bengal, 700053, India
| | - Soumi Das
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Arpan Adak
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata - Group of Institutions, Kolkata, West Bengal, 700053, India
| | - Swarupananda Mukherjee
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata - Group of Institutions, Kolkata, West Bengal, 700053, India.
| |
Collapse
|
6
|
Boonhok R, Senghoi W, Sangkanu S, Lim CL, Pudla M, Pereira MDL, Wilairatana P, Mahboob T, Rahman MA, Utaisincharoen P, Hiransai P, Nissapatorn V. Acanthamoeba castellanii-Mediated Reduction of Interleukin-1β Secretion and Its Association With Macrophage Autophagy. SCIENTIFICA 2025; 2025:3430892. [PMID: 40109888 PMCID: PMC11922611 DOI: 10.1155/sci5/3430892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 02/21/2025] [Indexed: 03/22/2025]
Abstract
Noncanonical autophagy including unconventional protein secretion has been extensively studied. Our work focused on a leaderless IL-1β protein secretion from human macrophage in response to Acanthamoeba castellanii components, Acanthamoeba culture supernatant (CS) and cell lysate (CL), as well as its association with macrophage autophagy. Phorbol 12-myristate 13-acetate (PMA)-induced THP-1 macrophages were treated with Acanthamoeba components of pathogenic (ATCC50739) and nonpathogenic (ATCC30010) strains in vitro. The data showed that Acanthamoeba treatment resulted in low IL-1β secretion from macrophages. In addition, Acanthamoeba CL of both strains was able to upregulate autophagy-related (Atg) protein 8, an autophagy marker, whereas Acanthamoeba CS downregulated Atg8 expression. We further manipulated autophagy and found that autophagy induction by starvation diminished IL-1β secretion while autophagy inhibition by 3-methyladenine (3MA) increased IL-1β secretion. Interestingly, in the presence of Acanthamoeba components either under starvation or 3MA treatment, IL-1β secretion was significantly reduced. Transcriptional expression of other ATG genes, i.e., ATG6, ATG7, and ATG5, were investigated and showed that their mRNA expression was maintained at the basal level under A. castellanii CS or CL treatment. Inflammasome-related genes, NLRP3 and CASPASE1, were upregulated following A. castellanii 50739 CS treatment but not in A. castellanii 50739 CL-treated condition. However, both conditions were able to increase IL-1β mRNA expression. TEM micrographs revealed that 3MA treatment induced the formation of large vacuoles and accumulation of autophagosome at the edge of THP-1 macrophages. However, the number and size of their structures were declined in the presence of A. castellanii 50739 CS with 3MA. Furthermore, immunofluorescence staining demonstrated the association between Atg8/LC3 and IL-1β expression, where downregulation of Atg8 by A. castellanii 50739 CS led to the upregulation of IL-1β. Altogether, the data indicate that Acanthamoeba can manipulate macrophage autophagy, thereby controlling low IL-1β secretion. The expression of autophagy- and inflammasome-related genes also indicates multiple mechanisms in IL-1β secretion in response to Acanthamoeba components. However, further characterization of Atg proteins and investigations into other intracellular pathways or defense mechanisms are needed to fully understand the unconventional secretion of IL-1β in macrophages. This knowledge could eventually lead to the development of innovative therapeutic strategies against Acanthamoeba infection by modulating autophagy or macrophage responses.
Collapse
Affiliation(s)
- Rachasak Boonhok
- Department of Medical Technology, School of Allied Health Sciences, and Research Excellence Center for Innovation and Health Products (RECIHP), Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Wilaiwan Senghoi
- Department of Medical Technology, School of Allied Health Sciences, and Center of Excellence Research for Melioidosis and Microorganisms (CERMM), Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Suthinee Sangkanu
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Songkhla 90112, Thailand
| | - Chooi Ling Lim
- Division of Applied Biomedical Science and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Matsayapan Pudla
- Department of Oral Microbiology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand
| | - Maria de Lourdes Pereira
- CICECO-Aveiro Institute of Materials and Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Tooba Mahboob
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | - Md Atiar Rahman
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong 4331, Bangladesh
| | - Pongsak Utaisincharoen
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Poonsit Hiransai
- Department of Medical Technology, School of Allied Health Sciences, and Center of Excellence in Marijuana, Hemp, and Kratom, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Veeranoot Nissapatorn
- School of Allied Health Sciences, Southeast Asia Water Team (SEA Water Team) and World Union for Herbal Drug Discovery (WUHeDD), Walailak University, Nakhon Si Thammarat 80160, Thailand
| |
Collapse
|
7
|
Valério-Bolas A, Meunier M, Rodrigues A, Palma-Marques J, Ferreira R, Cardoso I, Lobo L, Monteiro M, Nunes T, Armada A, Antunes WT, Alexandre-Pires G, da Fonseca IP, Santos-Gomes G. Unveiling the Interplay Between Dendritic Cells and Natural Killer Cells as Key Players in Leishmania Infection. J Immunol Res 2025; 2025:3176927. [PMID: 39963187 PMCID: PMC11832263 DOI: 10.1155/jimr/3176927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 01/02/2025] [Indexed: 02/20/2025] Open
Abstract
Leishmaniasis is a group of parasitic diseases whose etiological agent is the protozoa Leishmania. These diseases afflict impoverished populations in tropical and subtropical regions and affect wild and domestic animals. Canine leishmaniasis is a global disease mostly caused by L. infantum. Dogs are recognized as a good reservoir since harbor the infection long before developing the disease, facilitating parasite transmission. Furthermore, there is growing evidence that dogs may also be the reservoir of the American Leishmania spp. as L. amazonensis. The innate immune response is the first defense line against pathogens, which includes natural killer (NK) and dendritic cells (DCs). By recognizing and ultimately destroying infected cells, and by secreting immune mediators that favor inflammatory microenvironments, NK cells take the lead in the infectious process. When interacting with Leishmania parasites, DCs become activated and play a key role in driving the host immune response. While activated DCs can modulate NK cell activity, Leishmania parasites can directly activate NK cells by interacting with innate immune receptors. Once activated, NK cells can engage in a bidirectional interplay with DCs. However, the complexity of these interactions during Leishmania infection makes it challenging to fully understand the underlying processes. To further explore this, the present study investigated the dynamic interplay established between monocyte-derived DCs (moDCs) and putative NK (pNK) cells of dogs during Leishmania infection. Findings indicate that the crosstalk between moDCs exposed to L. infantum or L. amazonensis and pNK cells enhances chemokine upregulation, potentially attracting other leukocytes to the site of infection. pNK cells activated by L. infantum infected DCs upregulate IL-10, which can lead to a regulatory immune response while moDCs exposed to L. amazonensis induced pNK cells to overexpress IFN-γ and IL-13, favoring a mix of pro- and anti-inflammatory response. In addition, parasite-derived extracellular vesicles (EVs) can modulate the host immune response by stimulating the upregulation of anti-inflammatory cytokines and perforin release, which may impact infection outcomes. Thus, Leishmania and parasitic EVs can influence the bidirectional interplay between canine NK cells and DCs.
Collapse
Affiliation(s)
- Ana Valério-Bolas
- Unit for Teaching and Research in Medical Parasitology, Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Lisbon 1349-008, Portugal
| | - Mafalda Meunier
- Unit for Teaching and Research in Medical Parasitology, Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Lisbon 1349-008, Portugal
| | - Armanda Rodrigues
- Unit for Teaching and Research in Medical Parasitology, Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Lisbon 1349-008, Portugal
| | - Joana Palma-Marques
- Unit for Teaching and Research in Medical Parasitology, Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Lisbon 1349-008, Portugal
| | - Rui Ferreira
- BSA, Banco de Sangue Animal, Porto 4100-462, Portugal
| | - Inês Cardoso
- BSA, Banco de Sangue Animal, Porto 4100-462, Portugal
| | - Lis Lobo
- Unit for Teaching and Research in Medical Parasitology, Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Lisbon 1349-008, Portugal
| | - Marta Monteiro
- Unit for Teaching and Research in Medical Parasitology, Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Lisbon 1349-008, Portugal
- Faculty of Veterinary Medicine, Centre for Interdisciplinary Research in Animal Health, CIISA, University of Lisbon, Av. Universidade Técnica, Lisbon 1300-477, Portugal
| | - Telmo Nunes
- Microscopy Center, Faculty of Sciences of the University of Lisbon-FCUL—BioISI Ce3CE, Lisboa, Portugal
| | - Ana Armada
- Unit for Teaching and Research in Medical Parasitology, Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Lisbon 1349-008, Portugal
| | - Wilson T. Antunes
- Instituto Universitário Militar (IUM), Centro de Investigação, Desenvolvimento e Inovação da Academia Militar (CINAMIL), Unidade Militar Laboratorial de Defesa Biológica e Química (UMLDBQ), Lisboa 1849-012, Portugal
| | - Graça Alexandre-Pires
- Faculty of Veterinary Medicine, Centre for Interdisciplinary Research in Animal Health, CIISA, University of Lisbon, Av. Universidade Técnica, Lisbon 1300-477, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Isabel Pereira da Fonseca
- Faculty of Veterinary Medicine, Centre for Interdisciplinary Research in Animal Health, CIISA, University of Lisbon, Av. Universidade Técnica, Lisbon 1300-477, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Gabriela Santos-Gomes
- Unit for Teaching and Research in Medical Parasitology, Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Lisbon 1349-008, Portugal
| |
Collapse
|
8
|
Kratzer B, Gattinger P, Trapin D, Ettel P, Körmöczi U, Rottal A, Stieger RB, Sehgal ANA, Feichter M, Borochova K, Tulaeva I, Grabmeier-Pfistershammer K, Tauber PA, Perkmann T, Fae I, Wenda S, Kundi M, Fischer GF, Valenta R, Pickl WF. Differential decline of SARS-CoV-2-specific antibody levels, innate and adaptive immune cells, and shift of Th1/inflammatory to Th2 serum cytokine levels long after first COVID-19. Allergy 2024; 79:2482-2501. [PMID: 39003594 DOI: 10.1111/all.16210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/23/2024] [Accepted: 05/25/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND SARS-CoV-2 has triggered a pandemic and contributes to long-lasting morbidity. Several studies have investigated immediate cellular and humoral immune responses during acute infection. However, little is known about long-term effects of COVID-19 on the immune system. METHODS We performed a longitudinal investigation of cellular and humoral immune parameters in 106 non-vaccinated subjects ten weeks (10 w) and ten months (10 m) after their first SARS-CoV-2 infection. Peripheral blood immune cells were analyzed by multiparametric flow cytometry, serum cytokines were examined by multiplex technology. Antibodies specific for the Spike protein (S), the receptor-binding domain (RBD) and the nucleocapsid protein (NC) were determined. All parameters measured 10 w and 10 m after infection were compared with those of a matched, noninfected control group (n = 98). RESULTS Whole blood flow cytometric analyses revealed that 10 m after COVID-19, convalescent patients compared to controls had reduced absolute granulocyte, monocyte, and lymphocyte counts, involving T, B, and NK cells, in particular CD3+CD45RA+CD62L+CD31+ recent thymic emigrant T cells and non-class-switched CD19+IgD+CD27+ memory B cells. Cellular changes were associated with a reversal from Th1- to Th2-dominated serum cytokine patterns. Strong declines of NC- and S-specific antibody levels were associated with younger age (by 10.3 years, p < .01) and fewer CD3-CD56+ NK and CD19+CD27+ B memory cells. Changes of T-cell subsets at 10 m such as normalization of effector and Treg numbers, decline of RTE, and increase of central memory T cell numbers were independent of antibody decline pattern. CONCLUSIONS COVID-19 causes long-term reduction of innate and adaptive immune cells which is associated with a Th2 serum cytokine profile. This may provide an immunological mechanism for long-term sequelae after COVID-19.
Collapse
Affiliation(s)
- Bernhard Kratzer
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Pia Gattinger
- Center for Pathophysiology, Infectiology and Immunology, Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Doris Trapin
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Paul Ettel
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Ulrike Körmöczi
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Arno Rottal
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Robert B Stieger
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Al Nasar Ahmed Sehgal
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Melanie Feichter
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Kristina Borochova
- Center for Pathophysiology, Infectiology and Immunology, Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Inna Tulaeva
- Center for Pathophysiology, Infectiology and Immunology, Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | | | - Peter A Tauber
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Thomas Perkmann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Ingrid Fae
- Department of Transfusion Medicine and Cell Therapy, Medical University of Vienna, Vienna, Austria
| | - Sabine Wenda
- Department of Transfusion Medicine and Cell Therapy, Medical University of Vienna, Vienna, Austria
| | - Michael Kundi
- Center for Public Health, Department for Environmental Health, Medical University of Vienna, Vienna, Austria
| | - Gottfried F Fischer
- Department of Transfusion Medicine and Cell Therapy, Medical University of Vienna, Vienna, Austria
| | - Rudolf Valenta
- Center for Pathophysiology, Infectiology and Immunology, Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- NRC Institute of Immunology FMBA of Russia, Moscow, Russia
- Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Winfried F Pickl
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
- Karl Landsteiner University of Health Sciences, Krems, Austria
| |
Collapse
|
9
|
Dotta E, Maciola AK, Baccega T, Pasqual G. Dendritic cells steering antigen and leukocyte traffic in lymph nodes. FEBS Lett 2024. [PMID: 38997244 DOI: 10.1002/1873-3468.14982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 06/01/2024] [Accepted: 06/11/2024] [Indexed: 07/14/2024]
Abstract
Dendritic cells (DCs) play a central role in initiating and shaping the adaptive immune response, thanks to their ability to uptake antigens and present them to T cells. Once in the lymph node (LN), DCs can spread the antigen to other DCs, expanding the pool of cells capable of activating specific T-cell clones. Additionally, DCs can modulate the dynamics of other immune cells, by increasing naïve T-cell dwell time, thereby facilitating the scanning for cognate antigens, and by selectively recruiting other leukocytes. Here we discuss the role of DCs in orchestrating antigen and leukocyte trafficking within the LN, together with the implications of this trafficking on T-cell activation and commitment to effector function.
Collapse
Affiliation(s)
- Enrico Dotta
- Laboratory of Synthetic Immunology, Oncology and Immunology Section, Department of Surgery Oncology and Gastroenterology, University of Padua, Italy
| | - Agnieszka Katarzyna Maciola
- Laboratory of Synthetic Immunology, Oncology and Immunology Section, Department of Surgery Oncology and Gastroenterology, University of Padua, Italy
| | - Tania Baccega
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Giulia Pasqual
- Laboratory of Synthetic Immunology, Oncology and Immunology Section, Department of Surgery Oncology and Gastroenterology, University of Padua, Italy
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| |
Collapse
|
10
|
Lawrence LA, Vidal P, Varughese RS, Tiger Li ZR, Chen TD, Tuske SC, Jimenez AR, Lowen AC, Shafer WM, Swaims-Kohlmeier A. Murine modeling of menstruation identifies immune correlates of protection during Chlamydia muridarum challenge. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595090. [PMID: 38826233 PMCID: PMC11142139 DOI: 10.1101/2024.05.21.595090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
The menstrual cycle influences the risk of acquiring sexually transmitted infections (STIs), including Chlamydia trachomatis (C. trachomatis), although the underlying immune contributions are poorly defined. A mouse model simulating the immune-mediated process of menstruation could provide valuable insights into tissue-specific determinants of protection against chlamydial infection within the cervicovaginal and uterine mucosae comprising the female reproductive tract (FRT). Here, we used the pseudopregnancy approach in naïve C57Bl/6 mice and performed vaginal challenge with Chlamydia muridarum (C. muridarum) at decidualization, endometrial tissue remodeling, or uterine repair. This strategy identified that the time frame comprising uterine repair correlated with robust infection and greater bacterial burden as compared with mice on hormonal contraception, while challenges during endometrial remodeling were least likely to result in a productive infection. By comparing the infection site at early time points following chlamydial challenge, we found that a greater abundance of innate effector populations and proinflammatory signaling, including IFNγ correlated with protection. FRT immune profiling in uninfected mice over pseudopregnancy or in pig-tailed macaques over the menstrual cycle identified NK cell infiltration into the cervicovaginal tissues and lumen over the course of endometrial remodeling. Notably, NK cell depletion over this time frame reversed protection, with mice now productively infected with C. muridarum following challenge. This study shows that the pseudopregnancy murine menstruation model recapitulates immune changes in the FRT as a result of endometrial remodeling and identifies NK cell localization at the FRT as essential for immune protection against primary C. muridarum infection.
Collapse
Affiliation(s)
- Laurel A Lawrence
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
| | - Paola Vidal
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
| | - Richa S Varughese
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
| | - Zheng-Rong Tiger Li
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
| | - Thien Duy Chen
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
| | - Steven C Tuske
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
| | - Ariana R Jimenez
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
| | - Anice C Lowen
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
| | - William M Shafer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Laboratories of Bacterial Pathogenesis, Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - Alison Swaims-Kohlmeier
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Department of GYNOB, Emory University School of Medicine, Atlanta, Georgia
- Division of HIV Prevention Centers for Disease Control and Prevention, Atlanta, Georgia (previous affiliation)
| |
Collapse
|
11
|
Hong GH, Lee SY, Kim IA, Suk J, Baeg C, Kim JY, Lee S, Kim KJ, Kim KT, Kim MG, Park KY. Effect of Heat-Treated Lactiplantibacillus plantarum nF1 on the Immune System Including Natural Killer Cell Activity: A Randomized, Placebo-Controlled, Double-Blind Study. Nutrients 2024; 16:1339. [PMID: 38732587 PMCID: PMC11085399 DOI: 10.3390/nu16091339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 04/26/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024] Open
Abstract
Heat-treated Lactiplantibacillus plantarum nF1 (HT-nF1) increases immune cell activation and the production of various immunomodulators (e.g., interleukin (IL)-12) as well as immunoglobulin (Ig) G, which plays an important role in humoral immunity, and IgA, which activates mucosal immunity. To determine the effect of HT-nF1 intake on improving immune function, a randomized, double-blind, placebo-controlled study was conducted on 100 subjects with normal white blood cell counts. The HT-nF1 group was administered capsules containing 5 × 1011 cells of HT-nF1 once a day for 8 weeks. After 8 weeks of HT-nF1 intake, significant changes in IL-12 were observed in the HT-nF1 group (p = 0.045). In particular, the change in natural killer (NK) cell activity significantly increased in subjects with low secretory (s) IgA (≤49.61 μg/mL) and low NK activity (E:T = 10:1) (≤3.59%). These results suggest that HT-nF1 has no safety issues and improves the innate immune function by regulating T helper (Th)1-related immune factors. Therefore, we confirmed that HT-nF1 not only has a positive effect on regulating the body's immunity, but it is also a safe material for the human body, which confirms its potential as a functional health food ingredient.
Collapse
Affiliation(s)
- Geun-Hye Hong
- IMMUNOBIOTECH Corp., Seoul 06628, Republic of Korea; (G.-H.H.); (S.-Y.L.)
| | - So-Young Lee
- IMMUNOBIOTECH Corp., Seoul 06628, Republic of Korea; (G.-H.H.); (S.-Y.L.)
| | - In Ah Kim
- Global Medical Research Center, Seoul 03737, Republic of Korea; (I.A.K.); (J.S.); (C.B.)
| | - Jangmi Suk
- Global Medical Research Center, Seoul 03737, Republic of Korea; (I.A.K.); (J.S.); (C.B.)
| | - Chaemin Baeg
- Global Medical Research Center, Seoul 03737, Republic of Korea; (I.A.K.); (J.S.); (C.B.)
| | - Ji Yeon Kim
- Department of Food Science and Biotechnology, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea; (J.Y.K.); (S.L.)
| | - Sehee Lee
- Department of Food Science and Biotechnology, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea; (J.Y.K.); (S.L.)
| | - Kyeong Jin Kim
- Department of Nano Bio Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea;
| | - Ki Tae Kim
- IMMUNOBIOTECH Corp., Seoul 06628, Republic of Korea; (G.-H.H.); (S.-Y.L.)
| | - Min Gee Kim
- IMMUNOBIOTECH Corp., Seoul 06628, Republic of Korea; (G.-H.H.); (S.-Y.L.)
| | - Kun-Young Park
- IMMUNOBIOTECH Corp., Seoul 06628, Republic of Korea; (G.-H.H.); (S.-Y.L.)
| |
Collapse
|
12
|
Mulè A, Crosato V, Kuhns DB, Lorenzi L, Chirico C, Maifredi G, Notarangelo LD, Castelli F, Tomasoni LR. Visceral Leishmaniasis in Immunocompetent Hosts in Brescia: A Case Series and Analysis of Cytokine Cascade. Microorganisms 2024; 12:394. [PMID: 38399799 PMCID: PMC10892745 DOI: 10.3390/microorganisms12020394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Visceral leishmaniasis (VL) is a parasitic zoonosis caused by Leishmania spp. that usually manifests itself in immunocompromised subjects. It is a rare and neglected disease, and it is not endemic in the province of Brescia (Italy). Three cases of human VL occurred in Brescia from October to December 2021 in immunocompetent patients. We evaluated the patients looking for signs of underlying immunodeficiencies and conducted further epidemiological evaluations in the province of Brescia without success. An analysis of the sera levels of the main cytokines involved in the immune response to VL was performed. All patients presented a significant augmentation of CXCL-10, CCL-4, and IL-6. The patients tested during the acute phase showed an elevation of IL-1α, IL-5, IL-10, and IL-12, while in the recovery phase, higher levels of TNF-α and IL-7 were detected. Altogether, a predominant activation of the T-helper-2 pathway emerged during the acute phase of the parasite infection, while the cytokines associated with the T-helper-1 pathway were less represented. This imbalanced immune response to the parasite infection might play a crucial role in the development of VL in immunocompetent patients.
Collapse
Affiliation(s)
- Alice Mulè
- Unit of Infectious and Tropical Diseases, Department of Clinical and Experimental Sciences, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (V.C.); (F.C.)
| | - Verena Crosato
- Unit of Infectious and Tropical Diseases, Department of Clinical and Experimental Sciences, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (V.C.); (F.C.)
| | - Douglas Byron Kuhns
- Neutrophil Monitoring Laboratory, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 20701, USA;
| | - Luisa Lorenzi
- Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy;
| | - Claudia Chirico
- Department of Hygiene and Health Prevention, Health Protection Agency of Brescia (ATS Brescia), 25124 Brescia, Italy
| | - Giovanni Maifredi
- Epidemiology Unit, Health Protection Agency of Brescia (ATS Brescia), 25124 Brescia, Italy;
| | - Luigi D. Notarangelo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, NIAID, NIH, Bethesda, MD 20852, USA;
| | - Francesco Castelli
- Unit of Infectious and Tropical Diseases, Department of Clinical and Experimental Sciences, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (V.C.); (F.C.)
| | - Lina R. Tomasoni
- Unit of Infectious and Tropical Diseases, ASST Spedali Civili di Brescia, 25123 Brescia, Italy;
| |
Collapse
|
13
|
Horta AL, Gigley J, Boutet M, Lavau G, Weiss LM, Huang H. Memory-like NK Cells Are a Critical Component of Vaccine-Induced Immunity to Trypanosoma cruzi Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:617-631. [PMID: 38197653 PMCID: PMC10872457 DOI: 10.4049/jimmunol.2300509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 12/06/2023] [Indexed: 01/11/2024]
Abstract
Chagas disease by Trypanosoma cruzi infection is a major public health issue. The available therapeutic agents have limited efficacy and significant side effects. A reliable vaccine would reduce the threat of T. cruzi infections and prevent Chagas disease. Understanding the immune response to this infection would improve vaccine design. We previously demonstrated that adoptively transferred NK cells from mice immunized with highly attenuated T. cruzi, GFP-DDDHA strain, provided potent protection in naive recipients against secondary lethal challenge with various wild-type (WT) strains. To understand the importance of NK cells in protecting mice against T. cruzi infection, we performed an in-depth characterization of NK cell phenotype, responses, and memory-like traits during acute infections due to GFP-DDDHA and WT strains and in immunized mice during a recall response to a WT lethal challenge. NK cells robustly expanded and became more mature and cytolytic during the GFP-DDDHA strain immunization. NK cells in immunized mice responded more robustly after WT lethal challenge than during an acute primary WT infection. In addition, protection by immunization with the GFP-DDDHA strain is significantly weakened in NK cell-deficient mice and did not prevent parasitemia from WT lethal challenge, indicating that NK cells with memory-like traits were a critical component for early control of WT lethal challenge. Prior T. cruzi vaccine development studies have not included studies of this rapid NK response. These findings provide insights into overcoming existing challenges in developing a safe and effective vaccine to prevent this infection.
Collapse
Affiliation(s)
- Aline L. Horta
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Jason Gigley
- Department of Molecular Biology, University of Wyoming, Laramie, Wyoming, United States of America
| | - Marie Boutet
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Gregoire Lavau
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Louis M. Weiss
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Huan Huang
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| |
Collapse
|
14
|
Ramos A, Koch CE, Liu-Lupo Y, Hellinger RD, Kyung T, Abbott KL, Fröse J, Goulet D, Gordon KS, Eidell KP, Leclerc P, Whittaker CA, Larson RC, Muscato AJ, Yates KB, Dubrot J, Doench JG, Regev A, Vander Heiden MG, Maus MV, Manguso RT, Birnbaum ME, Hemann MT. Leukemia-intrinsic determinants of CAR-T response revealed by iterative in vivo genome-wide CRISPR screening. Nat Commun 2023; 14:8048. [PMID: 38052854 PMCID: PMC10698189 DOI: 10.1038/s41467-023-43790-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/10/2023] [Indexed: 12/07/2023] Open
Abstract
CAR-T therapy is a promising, novel treatment modality for B-cell malignancies and yet many patients relapse through a variety of means, including loss of CAR-T cells and antigen escape. To investigate leukemia-intrinsic CAR-T resistance mechanisms, we performed genome-wide CRISPR-Cas9 loss-of-function screens in an immunocompetent murine model of B-cell acute lymphoblastic leukemia (B-ALL) utilizing a modular guide RNA library. We identified IFNγR/JAK/STAT signaling and components of antigen processing and presentation pathway as key mediators of resistance to CAR-T therapy in vivo; intriguingly, loss of this pathway yielded the opposite effect in vitro (sensitized leukemia to CAR-T cells). Transcriptional characterization of this model demonstrated upregulation of these pathways in tumors relapsed after CAR-T treatment, and functional studies showed a surprising role for natural killer (NK) cells in engaging this resistance program. Finally, examination of data from B-ALL patients treated with CAR-T revealed an association between poor outcomes and increased expression of JAK/STAT and MHC-I in leukemia cells. Overall, our data identify an unexpected mechanism of resistance to CAR-T therapy in which tumor cell interaction with the in vivo tumor microenvironment, including NK cells, induces expression of an adaptive, therapy-induced, T-cell resistance program in tumor cells.
Collapse
Affiliation(s)
- Azucena Ramos
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Catherine E Koch
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yunpeng Liu-Lupo
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Riley D Hellinger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Taeyoon Kyung
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Keene L Abbott
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Julia Fröse
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Daniel Goulet
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Khloe S Gordon
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Keith P Eidell
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Paul Leclerc
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Charles A Whittaker
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rebecca C Larson
- Cellular Immunotherapy Program, Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Immunology Program, Harvard Medical School, Boston, MA, USA
| | - Audrey J Muscato
- Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, 02142, USA
| | - Kathleen B Yates
- Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, 02142, USA
| | - Juan Dubrot
- Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, 02142, USA
- Solid Tumors Program, Division of Oncology, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - John G Doench
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, 02142, USA
| | - Aviv Regev
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, 02142, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Immunology Program, Harvard Medical School, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, 02142, USA
- Ragon Institute of MIT, MGH, and Harvard, Cambridge, MA, USA
| | - Robert T Manguso
- Immunology Program, Harvard Medical School, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, 02142, USA
| | - Michael E Birnbaum
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Michael T Hemann
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
15
|
Cao W, Luo C, Fan Z, Lei M, Cheng X, Shi Z, Mao F, Xu Q, Fu Z, Zhang Q. Analysis of potential biomarkers and immune infiltration in autism based on bioinformatics analysis. Medicine (Baltimore) 2023; 102:e33340. [PMID: 37171362 PMCID: PMC10174422 DOI: 10.1097/md.0000000000033340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 05/13/2023] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder caused by both environmental and genetic factors. However, its etiology and pathogenesis remain unclear. The purpose of this study was to establish an immune-related diagnostic model for ASD using bioinformatics methods and to identify ASD biomarkers. Two ASD datasets, GSE18123 and GSE29691, were integrated into the gene expression Database to eliminate batch effects. 41 differentially expressed genes were identified by microarray data linear model (limma package). Based on the results of the immune infiltration analysis, we speculated that neutrophils, B cells naive, CD8+ T cells, and Tregs are potential core immune cells in ASD and participate in the occurrence of ASD. Finally, the differential genes and immune infiltration in ASD and non-ASD patients were compared, and the most relevant genes were selected to construct the first immune correlation prediction model of ASD. After the calculation, the model exhibited better accuracy. The calculations show that the model has good accuracy.
Collapse
Affiliation(s)
- Wenjun Cao
- Neonatal Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Clinical Treatment and Follow-up Center for High-risk Newborns of Henan Province, Zhengzhou, China
- Key Laboratory for Prevention and Control of Developmental Disorders, Zhengzhou, China
| | - Chenghan Luo
- Orthopeadics Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhaohan Fan
- National Engineering Laboratory for Internet Medical Systems and Applications, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengyuan Lei
- Health Care Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinru Cheng
- Neonatal Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Clinical Treatment and Follow-up Center for High-risk Newborns of Henan Province, Zhengzhou, China
- Key Laboratory for Prevention and Control of Developmental Disorders, Zhengzhou, China
| | - Zanyang Shi
- Neonatal Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Clinical Treatment and Follow-up Center for High-risk Newborns of Henan Province, Zhengzhou, China
- Key Laboratory for Prevention and Control of Developmental Disorders, Zhengzhou, China
| | - Fengxia Mao
- Neonatal Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Clinical Treatment and Follow-up Center for High-risk Newborns of Henan Province, Zhengzhou, China
| | - Qianya Xu
- Neonatal Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Clinical Treatment and Follow-up Center for High-risk Newborns of Henan Province, Zhengzhou, China
| | - Zhaoqin Fu
- Neonatal Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Clinical Treatment and Follow-up Center for High-risk Newborns of Henan Province, Zhengzhou, China
| | - Qian Zhang
- Neonatal Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Clinical Treatment and Follow-up Center for High-risk Newborns of Henan Province, Zhengzhou, China
- Key Laboratory for Prevention and Control of Developmental Disorders, Zhengzhou, China
| |
Collapse
|
16
|
Roles of natural killer cells in immunity to cancer, and applications to immunotherapy. Nat Rev Immunol 2023; 23:90-105. [PMID: 35637393 DOI: 10.1038/s41577-022-00732-1] [Citation(s) in RCA: 256] [Impact Index Per Article: 128.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2022] [Indexed: 02/04/2023]
Abstract
Great strides have been made in recent years towards understanding the roles of natural killer (NK) cells in immunity to tumours and viruses. NK cells are cytotoxic innate lymphoid cells that produce inflammatory cytokines and chemokines. By lysing transformed or infected cells, they limit tumour growth and viral infections. Whereas T cells recognize peptides presented by MHC molecules, NK cells display receptors that recognize stress-induced autologous proteins on cancer cells. At the same time, their functional activity is inhibited by MHC molecules displayed on such cells. The enormous potential of NK cells for immunotherapy for cancer is illustrated by their broad recognition of stressed cells regardless of neoantigen presentation, and enhanced activity against tumours that have lost expression of MHC class I owing to acquired resistance mechanisms. As a result, many efforts are under way to mobilize endogenous NK cells with therapeutics, or to provide populations of ex vivo-expanded NK cells as a cellular therapy, in some cases by equipping the NK cells with chimeric antigen receptors. Here we consider the key features that underlie why NK cells are emerging as important new additions to the cancer therapeutic arsenal.
Collapse
|
17
|
Genetic Iron Overload Hampers Development of Cutaneous Leishmaniasis in Mice. Int J Mol Sci 2023; 24:ijms24021669. [PMID: 36675185 PMCID: PMC9864902 DOI: 10.3390/ijms24021669] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
The survival, growth, and virulence of Leishmania spp., a group of protozoan parasites, depends on the proper access and regulation of iron. Macrophages, Leishmania's host cell, may divert iron traffic by reducing uptake or by increasing the efflux of iron via the exporter ferroportin. This parasite has adapted by inhibiting the synthesis and inducing the degradation of ferroportin. To study the role of iron in leishmaniasis, we employed Hjv-/- mice, a model of hemochromatosis. The disruption of hemojuvelin (Hjv) abrogates the expression of the iron hormone hepcidin. This allows unrestricted iron entry into the plasma from ferroportin-expressing intestinal epithelial cells and tissue macrophages, resulting in systemic iron overload. Mice were injected with Leishmania major in hind footpads or intraperitoneally. Compared with wild-type controls, Hjv-/- mice displayed transient delayed growth of L. major in hind footpads, with a significant difference in parasite burden 4 weeks post-infection. Following acute intraperitoneal exposure to L. major, Hjv-/- peritoneal cells manifested increased expression of inflammatory cytokines and chemokines (Il1b, Tnfa, Cxcl2, and Ccl2). In response to infection with L. infantum, the causative agent of visceral leishmaniasis, Hjv-/- and control mice developed similar liver and splenic parasite burden despite vastly different tissue iron content and ferroportin expression. Thus, genetic iron overload due to hemojuvelin deficiency appears to mitigate the early development of only cutaneous leishmaniasis.
Collapse
|
18
|
Oh BLZ, Chan LWY, Chai LYA. Manipulating NK cellular therapy from cancer to invasive fungal infection: promises and challenges. Front Immunol 2023; 13:1044946. [PMID: 36969979 PMCID: PMC10034767 DOI: 10.3389/fimmu.2022.1044946] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/09/2022] [Indexed: 01/13/2023] Open
Abstract
The ideal strategy to fight an infection involves both (i) weakening the invading pathogen through conventional antimicrobial therapy, and (ii) strengthening defense through the augmentation of host immunity. This is even more pertinent in the context of invasive fungal infections whereby the majority of patients have altered immunity and are unable to mount an appropriate host response against the pathogen. Natural killer (NK) cells fit the requirement of an efficient, innate executioner of both tumour cells and pathogens – their unique, targeted cell killing mechanism, combined with other arms of the immune system, make them potent effectors. These characteristics, together with their ready availability (given the various sources of extrinsic NK cells available for harvesting), make NK cells an attractive choice as adoptive cellular therapy against fungi in invasive infections. Improved techniques in ex vivo NK cell activation with expansion, and more importantly, recent advances in genetic engineering including state-of-the-art chimeric antigen receptor platform development, have presented an opportune moment to harness this novel therapeutic as a key component of a multipronged strategy against invasive fungal infections.
Collapse
Affiliation(s)
- Bernice Ling Zhi Oh
- VIVA-University Children’s Cancer Centre, Khoo-Teck Puat-National University Children’s Medical Institute, National University Hospital, Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
| | - Louis Wei Yong Chan
- Clinician Scientist Academy, National University Health System, Singapore, Singapore
| | - Louis Yi Ann Chai
- Division of Infectious Diseases, Department of Medicine, National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore, Singapore
- *Correspondence: Louis Yi Ann Chai,
| |
Collapse
|
19
|
The Effective Treatment of Purpurin on Inflammation and Adjuvant-Induced Arthritis. Molecules 2023; 28:molecules28010366. [PMID: 36615560 PMCID: PMC9824476 DOI: 10.3390/molecules28010366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/24/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
Rubia cordifolia L. (Rubiaceae), one of the traditional anti-rheumatic herbal medicines in China, has been used to treat rheumatoid arthritis (RA) since ancient times. Purpurin, an active compound of Rubia cordifolia L., has been identified in previous studies and exerts antibacterial, antigenotoxic, anticancer, and antioxidant effects. However, the efficacy and the underlying mechanism of purpurin to alleviate RA are unclear. In this study, the effect of purpurin on inflammation was investigated using macrophage RAW264.7 inflammatory cells, induced by lipopolysaccharide (LPS), and adjuvant-induced arthritis (AIA) rat was established to explore the effect of purpurin on joint damage and immune disorders; the network pharmacology and molecular docking were integrated to dig out the prospective target. Purpurin showed significantly anti-inflammatory effect by reducing the content of IL-6, TNF-α, and IL-1β and increasing IL-10. Besides, purpurin obviously improved joint injury and hypotoxicity in the liver and spleen and regulated the level of FOXP3 and CD4+/CD8+. Furthermore, purpurin reduced the MMP3 content of AIA rats. Network pharmacology and molecular docking also suggested that MMP3 may be the key target of purpurin against RA. The results of this study strongly indicated that purpurin has a potential effect on anti-RA.
Collapse
|
20
|
Alizadeh Z, Omidnia P, Altalbawy FMA, Gabr GA, Obaid RF, Rostami N, Aslani S, Heidari A, Mohammadi H. Unraveling the role of natural killer cells in leishmaniasis. Int Immunopharmacol 2023; 114:109596. [PMID: 36700775 DOI: 10.1016/j.intimp.2022.109596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/07/2022] [Accepted: 12/11/2022] [Indexed: 12/24/2022]
Abstract
NK cells are known as frontline responders that are efficient in combating several maladies as well as leishmaniasis caused by Leishmania spp. As such they are being investigated to be used for adoptive transfer therapy and vaccine. In spite of the lack of antigen-specific receptors at their surface, NK cells can selectively recognize pathogens, accomplished by the activation of the receptors on the NK cell surface and also as the result of their effector functions. Activation of NK cells can occur through interaction between TLR-2 expressed on NK cells and. LPG of Leishmania parasites. In addition, NK cell activation can occur by cytokines (e.g., IFN-γ and IL-12) that also lead to producing cytokines and chemokines and lysis of target cells. This review summarizes several evidences that support NK cells activation for controlling leishmaniasis and the potentially lucrative roles of NK cells during leishmaniasis. Furthermore, we discuss strategies of Leishmania parasites in inhibiting NK cell functions. Leishmania LPG can utilizes TLR2 to evade host-immune responses. Also, Leishmania GP63 can directly binds to NK cells and modulates NK cell phenotype. Finally, this review analyzes the potentialities to harness NK cells effectiveness in therapy regimens and vaccinations.
Collapse
Affiliation(s)
- Zahra Alizadeh
- Department of Parasitology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Farag M A Altalbawy
- National Institute of Laser Enhanced Sciences (NILES), University of Cairo, Giza 12613, Egypt; Department of Chemistry, University College of Duba, University of Tabuk, Duba 71911, Saudi Arabia
| | - Gamal A Gabr
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; Agricultural Genetic Engineering Research Institute (AGERI), Agricultural Research Center, Giza, Egypt
| | - Rasha Fadhel Obaid
- Department of Biomedical Engineering, Al-Mustaqbal University College, Babylon, Iraq
| | - Narges Rostami
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Aslani
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Aliehsan Heidari
- Department of Parasitology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran; Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
21
|
Abdi K, Laky K, Abshari M, Hill EM, Lantz L, Singh NJ, Long EO. Dendritic cells Trigger IFN-γ secretion by NK cells independent of IL-12 and IL-18. Eur J Immunol 2022; 52:1431-1440. [PMID: 35816444 PMCID: PMC10608798 DOI: 10.1002/eji.202149733] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 06/09/2022] [Accepted: 07/07/2022] [Indexed: 11/10/2022]
Abstract
It is commonly believed that IL-12 produced by DCs in response to pathogens is the first signal that stimulates the production of IFN-γ by NK cells. However, IL-12 production by DCs in response to bacterial LPS depends on either engagement of CD40 by CD40L on activated T cells or IFN-γ from NK cells. This suggests that during the primary immune response, NK cells produce IFN-γ before IL-12 production by DCs. Here, using single-cell measurements, cell sorting and mouse lines deficient in IL-12, IL-23, type I IFN receptor and the IL-18 receptor, we show that a subset of BM-derived DCs characterized by low expression of MHC class II (MHCIIlow ) stimulates IFN-γ production by NK cells. The expression of Toll-like Receptor (TLR) 4 on DCs but not NK cells was required for such NK-derived IFN-γ. In addition, soluble factor(s) produced by LPS-activated MHCIIlow DCs were sufficient to induce IFN-γ production by NK cells independent of IL-12, IL-23, and IL-18. This response was enhanced in the presence of a low dose of IL-2. These results delineate a previously unknown pathway of DC-mediated IFN-γ production by NK cells, which is independent of commonly known cytokines.
Collapse
Affiliation(s)
- Kaveh Abdi
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA
| | - Karen Laky
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA
| | - Mehrnoosh Abshari
- National Institute of Dental and Craniofacial Research, National Institute of Health, Bethesda, MD, USA
| | - Elizabeth M. Hill
- Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Larry Lantz
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA
| | - Nevil J. Singh
- Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Eric O. Long
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA
| |
Collapse
|
22
|
Cavalcante MKDA, de Freitas e Silva R, Pereira VRA, Brelaz-de-Castro MCA. Opinion Article: NK Cells in Cutaneous Leishmaniasis: Protection or Damage? Front Immunol 2022; 13:933490. [PMID: 35844579 PMCID: PMC9283678 DOI: 10.3389/fimmu.2022.933490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 06/01/2022] [Indexed: 11/18/2022] Open
Affiliation(s)
- Marton Kaique de Andrade Cavalcante
- Department of Immunology, Aggeu Magalhães Institute - Oswaldo Cruz Foundation, Recife, Brazil
- Parasitology Laboratory, Federal University of Pernambuco, Vitoria de Santo Antão, Brazil
| | - Rafael de Freitas e Silva
- Department of Immunology, Aggeu Magalhães Institute - Oswaldo Cruz Foundation, Recife, Brazil
- Department of Natural Sciences, University of Pernambuco, Garanhuns, Brazil
| | | | - Maria Carolina Accioly Brelaz-de-Castro
- Department of Immunology, Aggeu Magalhães Institute - Oswaldo Cruz Foundation, Recife, Brazil
- Parasitology Laboratory, Federal University of Pernambuco, Vitoria de Santo Antão, Brazil
- *Correspondence: Maria Carolina Accioly Brelaz-de-Castro,
| |
Collapse
|
23
|
A century of attempts to develop an effective tuberculosis vaccine: Why they failed? Int Immunopharmacol 2022; 109:108791. [PMID: 35487086 DOI: 10.1016/j.intimp.2022.108791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/18/2022] [Accepted: 04/18/2022] [Indexed: 11/23/2022]
Abstract
Tuberculosis (TB) remains a major global health problem despite widespread use of the Bacillus BCG vaccine. This situation is worsened by co-infection with HIV, and the development of multidrug-resistant Mycobacterium tuberculosis (Mtb) strains. Thus, novel vaccine candidates and improved vaccination strategies are urgently needed in order to reduce the incidence of TB and even to eradicate TB by 2050. Over the last few decades, 23 novel TB vaccines have entered into clinical trials, more than 13 new vaccines have reached various stages of preclinical development, and more than 50 potential candidates are in the discovery stage as next-generation vaccines. Nevertheless, why has a century of attempts to introduce an effective TB vaccine failed? Who should be blamed -scientists, human response, or Mtb strategies? Literature review reveals that the elimination of latent or active Mtb infections in a given population seems to be an epigenetic process. With a better understanding of the connections between bacterial infections and gene expression conditions in epigenetic events, opportunities arise in designing protective vaccines or therapeutic agents, particularly as epigenetic processes can be reversed. Therefore, this review provides a brief overview of different approaches towards novel vaccination strategies and the mechanisms underlying these approaches.
Collapse
|
24
|
Immune Responses in Leishmaniases: An Overview. Trop Med Infect Dis 2022; 7:tropicalmed7040054. [PMID: 35448829 PMCID: PMC9029249 DOI: 10.3390/tropicalmed7040054] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
Leishmaniasis is a parasitic, widespread, and neglected disease that affects more than 90 countries in the world. More than 20 Leishmania species cause different forms of leishmaniasis that range in severity from cutaneous lesions to systemic infection. The diversity of leishmaniasis forms is due to the species of parasite, vector, environmental and social factors, genetic background, nutritional status, as well as immunocompetence of the host. Here, we discuss the role of the immune system, its molecules, and responses in the establishment, development, and outcome of Leishmaniasis, focusing on innate immune cells and Leishmania major interactions.
Collapse
|
25
|
Jafarzadeh A, Kumar S, Bodhale N, Jafarzadeh S, Nemati M, Sharifi I, Sarkar A, Saha B. The expression of PD-1 and its ligands increases in Leishmania infection and its blockade reduces the parasite burden. Cytokine 2022; 153:155839. [PMID: 35276636 DOI: 10.1016/j.cyto.2022.155839] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/16/2021] [Accepted: 02/21/2022] [Indexed: 11/03/2022]
Abstract
The expression of programmed cell death protein-1 (PD-1) and its ligands- PD-L1 and PD-L2- on T cells and macrophages', respectively, increases in Leishmania infection. The PD-1/PD-L1 interaction induces T cell anergy, T cell apoptosis and exhaustion, diversion of T cells toward TH2 and T-reg cells but inhibits M1 macrophage activities by suppression of nitric oxide (NO) and reactive oxygen species (ROS) production. These changes exacerbate Leishmania infection. As PD-L1-deficient, but not PD-L2-deficient, mice were protected againstL. mexicanainfection, differential roles have been proposed for PD-L1 and PD-L2 in mouse models of leishmaniasis. Blockade of PD-1/PD-L1 interaction in various in vitro and Leishmania-infected mouse, hamster and dog models enhanced IFN-γ and NO production, reduced IL-10 and TGF-β generation, promoted T cell proliferation and reduced parasite burden. Therefore, PD-1/PD-L1 blockade is being considered as a potential therapeutic strategy to restore protective immunity during leishmaniasis, particularly, in drug-resistant cases.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Sunil Kumar
- National Centre For Cell Science, Pune 411007, India
| | | | - Sara Jafarzadeh
- Student Research Committee, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Nemati
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Haematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Iraj Sharifi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Arup Sarkar
- Trident Academy of Creative Technology, Bhubaneswar, India
| | - Bhaskar Saha
- National Centre For Cell Science, Pune 411007, India; Student Research Committee, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Trident Academy of Creative Technology, Bhubaneswar, India.
| |
Collapse
|
26
|
Haegebaert RM, Kempers M, Ceelen W, Lentacker I, Remaut K. Nanoparticle mediated targeting of toll-like receptors to treat colorectal cancer. Eur J Pharm Biopharm 2022; 172:16-30. [PMID: 35074555 DOI: 10.1016/j.ejpb.2022.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/16/2021] [Accepted: 01/17/2022] [Indexed: 02/07/2023]
|
27
|
Angelo LS, Hogg GD, Abeynaike S, Bimler L, Vargas-Hernandez A, Paust S. Phenotypic and Functional Plasticity of CXCR6+ Peripheral Blood NK Cells. Front Immunol 2022; 12:810080. [PMID: 35173710 PMCID: PMC8841448 DOI: 10.3389/fimmu.2021.810080] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 12/28/2021] [Indexed: 12/23/2022] Open
Abstract
Human NK cells are comprised of phenotypic subsets, whose potentially unique functions remain largely unexplored. C-X-C-motif-chemokine-receptor-6 (CXCR6)+ NK cells have been identified as phenotypically immature tissue-resident NK cells in mice and humans. A small fraction of peripheral blood (PB)-NK cells also expresses CXCR6. However, prior reports about their phenotypic and functional plasticity are conflicting. In this study, we isolated, expanded, and phenotypically and functionally evaluated CXCR6+ and CXCR6– PB-NK cells, and contrasted results to bulk liver and spleen NK cells. We found that CXCR6+ and CXCR6– PB-NK cells preserved their distinct phenotypic profiles throughout 14 days of in vitro expansion (“day 14”), after which phenotypically immature CXCR6+ PB-NK cells became functionally equivalent to CXCR6– PB-NK cells. Despite a consistent reduction in CD16 expression and enhanced expression of the transcription factor Eomesodermin (Eomes), day 14 CXCR6+ PB-NK cells had superior antibody-dependent cellular cytotoxicity (ADCC) compared to CXCR6– PB-NK cells. Further, bulk liver NK cells responded to IL-15, but not IL-2 stimulation, with STAT-5 phosphorylation. In contrast, bulk splenic and PB-NK cells robustly responded to both cytokines. Our findings may allow for the selection of superior NK cell subsets for infusion products increasingly used to treat human diseases.
Collapse
Affiliation(s)
- Laura S. Angelo
- Center for Human Immunobiology, Department of Pediatrics, Texas Children’s Hospital, Houston, TX, United States
| | - Graham D. Hogg
- Center for Human Immunobiology, Department of Pediatrics, Texas Children’s Hospital, Houston, TX, United States
| | - Shawn Abeynaike
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States
| | - Lynn Bimler
- Center for Human Immunobiology, Department of Pediatrics, Texas Children’s Hospital, Houston, TX, United States
| | - Alexander Vargas-Hernandez
- Center for Human Immunobiology, Department of Pediatrics, Texas Children’s Hospital, Houston, TX, United States
| | - Silke Paust
- Center for Human Immunobiology, Department of Pediatrics, Texas Children’s Hospital, Houston, TX, United States
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States
- *Correspondence: Silke Paust,
| |
Collapse
|
28
|
Miller JS, Davis ZB, Helgeson E, Reilly C, Thorkelson A, Anderson J, Lima NS, Jorstad S, Hart GT, Lee JH, Safrit JT, Wong H, Cooley S, Gharu L, Chung H, Soon-Shiong P, Dobrowolski C, Fletcher CV, Karn J, Douek DC, Schacker TW. Safety and virologic impact of the IL-15 superagonist N-803 in people living with HIV: a phase 1 trial. Nat Med 2022; 28:392-400. [DOI: 10.1038/s41591-021-01651-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/30/2021] [Indexed: 12/26/2022]
|
29
|
Sliz A, Yokoyama WM. NK Cells and ILC1s in Cancer Biology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1365:41-55. [DOI: 10.1007/978-981-16-8387-9_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
30
|
Singh TP, Carvalho AM, Sacramento LA, Grice EA, Scott P. Microbiota instruct IL-17A-producing innate lymphoid cells to promote skin inflammation in cutaneous leishmaniasis. PLoS Pathog 2021; 17:e1009693. [PMID: 34699567 PMCID: PMC8570469 DOI: 10.1371/journal.ppat.1009693] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 11/05/2021] [Accepted: 10/12/2021] [Indexed: 12/26/2022] Open
Abstract
Innate lymphoid cells (ILCs) comprise a heterogeneous population of immune cells that maintain barrier function and can initiate a protective or pathological immune response upon infection. Here we show the involvement of IL-17A-producing ILCs in microbiota-driven immunopathology in cutaneous leishmaniasis. IL-17A-producing ILCs were RORγt+ and were enriched in Leishmania major infected skin, and topical colonization with Staphylococcus epidermidis before L. major infection exacerbated the skin inflammatory responses and IL-17A-producing RORγt+ ILC accumulation without impacting type 1 immune responses. IL-17A responses in ILCs were directed by Batf3 dependent CD103+ dendritic cells and IL-23. Moreover, experiments using Rag1-/- mice established that IL-17A+ ILCs were sufficient in driving the inflammatory responses as depletion of ILCs or neutralization of IL-17A diminished the microbiota mediated immunopathology. Taken together, this study indicates that the skin microbiota promotes RORγt+ IL-17A-producing ILCs, which augment the skin inflammation in cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Tej Pratap Singh
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (TPS); (PS)
| | - Augusto M. Carvalho
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Laís Amorim Sacramento
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Elizabeth A. Grice
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Phillip Scott
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (TPS); (PS)
| |
Collapse
|
31
|
Posttraumatic natural killer cell decrease is associated with septic complications. J Surg Res 2021; 270:94-103. [PMID: 34649071 DOI: 10.1016/j.jss.2021.08.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 05/22/2021] [Accepted: 08/27/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND There has been paucity in prospective studies investigating trauma-induced changes in the cellular immunity of HIV-seropositive patients and their impact on the clinical outcome after trauma surgery. The role of natural killer (NK) cells especially has not yet been fully elucidated, and the function of this lymphocyte subtype in the immune defense after trauma is still under debate. METHODS This prospective study included patients requiring surgery for abdominal gunshot wounds. A blood specimen was obtained on admission, 48 hours after the index operation and, in case of damage control surgery, 48 hours after the first planned second look operation. The quantity and changes of T-, B- and NK cells were analyzed via flow cytometry to investigate whether these numbers had an impact on the postoperative outcome. RESULTS A total of 62 patients were recruited in the analysis of which 38 were HIV-negative and 24 HIV-seropositive. After surgery, HIV-negative patients had a more severe decrease of their CD4+ T cells compared to the HIV-seropositive patients. Trauma resulted in a severe decrease of NK cells irrespective of the HIV-serostatus. Patients with more extensive NK cell drop had a significantly higher postoperative complication rate. CONCLUSIONS Our data support the association of trauma-induced NK cell decrease with a subsequent significantly higher rate of septic and surgical complications and suggest that these immune cells might play an important role in antibacterial immunity. Strengthening the NK cell function or limiting their decrease in the postoperative course might be of therapeutic value in severely injured trauma patients.
Collapse
|
32
|
Carneiro MB, Peters NC. The Paradox of a Phagosomal Lifestyle: How Innate Host Cell- Leishmania amazonensis Interactions Lead to a Progressive Chronic Disease. Front Immunol 2021; 12:728848. [PMID: 34557194 PMCID: PMC8452962 DOI: 10.3389/fimmu.2021.728848] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
Abstract
Intracellular phagosomal pathogens represent a formidable challenge for innate immune cells, as, paradoxically, these phagocytic cells can act as both host cells that support pathogen replication and, when properly activated, are the critical cells that mediate pathogen elimination. Infection by parasites of the Leishmania genus provides an excellent model organism to investigate this complex host-pathogen interaction. In this review we focus on the dynamics of Leishmania amazonensis infection and the host innate immune response, including the impact of the adaptive immune response on phagocytic host cell recruitment and activation. L. amazonensis infection represents an important public health problem in South America where, distinct from other Leishmania parasites, it has been associated with all three clinical forms of leishmaniasis in humans: cutaneous, muco-cutaneous and visceral. Experimental observations demonstrate that most experimental mouse strains are susceptible to L. amazonensis infection, including the C57BL/6 mouse, which is resistant to other species such as Leishmania major, Leishmania braziliensis and Leishmania infantum. In general, the CD4+ T helper (Th)1/Th2 paradigm does not sufficiently explain the progressive chronic disease established by L. amazonensis, as strong cell-mediated Th1 immunity, or a lack of Th2 immunity, does not provide protection as would be predicted. Recent findings in which the balance between Th1/Th2 immunity was found to influence permissive host cell availability via recruitment of inflammatory monocytes has also added to the complexity of the Th1/Th2 paradigm. In this review we discuss the roles played by innate cells starting from parasite recognition through to priming of the adaptive immune response. We highlight the relative importance of neutrophils, monocytes, dendritic cells and resident macrophages for the establishment and progressive nature of disease following L. amazonensis infection.
Collapse
Affiliation(s)
- Matheus B Carneiro
- Snyder Institute for Chronic Diseases, Departments of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine and Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Nathan C Peters
- Snyder Institute for Chronic Diseases, Departments of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine and Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
33
|
Khalil M, Wang D, Hashemi E, Terhune SS, Malarkannan S. Implications of a 'Third Signal' in NK Cells. Cells 2021; 10:cells10081955. [PMID: 34440725 PMCID: PMC8393955 DOI: 10.3390/cells10081955] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/17/2022] Open
Abstract
Innate and adaptive immune systems are evolutionarily divergent. Primary signaling in T and B cells depends on somatically rearranged clonotypic receptors. In contrast, NK cells use germline-encoded non-clonotypic receptors such as NCRs, NKG2D, and Ly49H. Proliferation and effector functions of T and B cells are dictated by unique peptide epitopes presented on MHC or soluble humoral antigens. However, in NK cells, the primary signals are mediated by self or viral proteins. Secondary signaling mediated by various cytokines is involved in metabolic reprogramming, proliferation, terminal maturation, or memory formation in both innate and adaptive lymphocytes. The family of common gamma (γc) cytokine receptors, including IL-2Rα/β/γ, IL-7Rα/γ, IL-15Rα/β/γ, and IL-21Rα/γ are the prime examples of these secondary signals. A distinct set of cytokine receptors mediate a ‘third’ set of signaling. These include IL-12Rβ1/β2, IL-18Rα/β, IL-23R, IL-27R (WSX-1/gp130), IL-35R (IL-12Rβ2/gp130), and IL-39R (IL-23Rα/gp130) that can prime, activate, and mediate effector functions in lymphocytes. The existence of the ‘third’ signal is known in both innate and adaptive lymphocytes. However, the necessity, context, and functional relevance of this ‘third signal’ in NK cells are elusive. Here, we define the current paradigm of the ‘third’ signal in NK cells and enumerate its clinical implications.
Collapse
Affiliation(s)
- Mohamed Khalil
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA; (M.K.); (D.W.); (E.H.)
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Dandan Wang
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA; (M.K.); (D.W.); (E.H.)
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Elaheh Hashemi
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA; (M.K.); (D.W.); (E.H.)
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Scott S. Terhune
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Correspondence: (S.S.T.); (S.M.)
| | - Subramaniam Malarkannan
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA; (M.K.); (D.W.); (E.H.)
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Correspondence: (S.S.T.); (S.M.)
| |
Collapse
|
34
|
Bhagyaraj E, Wang H, Yang X, Hoffman C, Akgul A, Goodwin ZI, Pascual DW. Mucosal Vaccination Primes NK Cell-Dependent Development of CD8 + T Cells Against Pulmonary Brucella Infection. Front Immunol 2021; 12:697953. [PMID: 34305935 PMCID: PMC8293993 DOI: 10.3389/fimmu.2021.697953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/21/2021] [Indexed: 11/13/2022] Open
Abstract
Past studies with the live, double-mutant B. abortus (znBAZ) strain resulted in nearly complete protection of mice against pulmonary challenge with wild-type (wt) Brucella via a dominant CD8+ T cell response. To understand the contribution innate immune cells in priming CD8+ T cell responses, mice were nasally dosed with wt B. abortus, smooth vaccine strain 19 (S19), or znBAZ, and examined for innate immune cell activation. Flow cytometric analysis revealed that znBAZ, but not wt B. abortus nor S19 infection, induces up to a 5-fold increase in the frequency of IFN-γ-producing NK cells in mouse lungs. These NK cells express increased CXCR3 and Ki67, indicating their recruitment and proliferation subsequent to znBAZ infection. Their activation status was augmented noted by the increased NKp46 and granzyme B, but decreased NKG2A expression. Further analysis demonstrated that both lung caspase-1+ inflammatory monocytes and monocyte-derived macrophages secrete chemokines and cytokines responsible for NK cell recruitment and activation. Moreover, neutralizing IL-18, an NK cell-activating cytokine, reduced the znBAZ-induced early NK cell response. NK cell depletion also significantly impaired lung dendritic cell (DC) activation and migration to the lower respiratory lymph nodes (LRLNs). Both lung DC activation and migration to LRLNs were significantly impaired in NK cell-depleted or IFN-γ-/- mice, particularly the CD11b+ and monocytic DC subsets. Furthermore, znBAZ vaccination significantly induced CD8+ T cells, and upon in vivo NK cell depletion, CD8+ T cells were reduced 3-fold compared to isotype-treated mice. In summary, these data show that znBAZ induces lung IFN-γ+ NK cells, which plays a critical role in influencing lung DC activation, migration, and promoting protective CD8+ T cell development.
Collapse
Affiliation(s)
| | | | | | | | | | | | - David W. Pascual
- Department of Infectious Diseases & Immunology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
35
|
Duygu B, Olieslagers TI, Groeneweg M, Voorter CEM, Wieten L. HLA Class I Molecules as Immune Checkpoints for NK Cell Alloreactivity and Anti-Viral Immunity in Kidney Transplantation. Front Immunol 2021; 12:680480. [PMID: 34295330 PMCID: PMC8290519 DOI: 10.3389/fimmu.2021.680480] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 06/14/2021] [Indexed: 12/12/2022] Open
Abstract
Natural killer (NK) cells are innate lymphocytes that can kill diseased- or virally-infected cells, mediate antibody dependent cytotoxicity and produce type I immune-associated cytokines upon activation. NK cells also contribute to the allo-immune response upon kidney transplantation either by promoting allograft rejection through lysis of cells of the transplanted organ or by promoting alloreactive T cells. In addition, they protect against viral infections upon transplantation which may be especially relevant in patients receiving high dose immune suppression. NK cell activation is tightly regulated through the integrated balance of signaling via inhibitory- and activating receptors. HLA class I molecules are critical regulators of NK cell activation through the interaction with inhibitory- as well as activating NK cell receptors, hence, HLA molecules act as critical immune checkpoints for NK cells. In the current review, we evaluate how NK cell alloreactivity and anti-viral immunity are regulated by NK cell receptors belonging to the KIR family and interacting with classical HLA class I molecules, or by NKG2A/C and LILRB1/KIR2DL4 engaging non-classical HLA-E or -G. In addition, we provide an overview of the methods to determine genetic variation in these receptors and their HLA ligands.
Collapse
Affiliation(s)
- Burcu Duygu
- Department of Transplantation Immunology, Maastricht University Medical Center, Maastricht, Netherlands.,GROW, School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| | - Timo I Olieslagers
- Department of Transplantation Immunology, Maastricht University Medical Center, Maastricht, Netherlands.,GROW, School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| | - Mathijs Groeneweg
- Department of Transplantation Immunology, Maastricht University Medical Center, Maastricht, Netherlands.,GROW, School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| | - Christina E M Voorter
- Department of Transplantation Immunology, Maastricht University Medical Center, Maastricht, Netherlands.,GROW, School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| | - Lotte Wieten
- Department of Transplantation Immunology, Maastricht University Medical Center, Maastricht, Netherlands.,GROW, School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
36
|
Kust SA, Streltsova MA, Panteleev AV, Karpina NL, Lyadova IV, Sapozhnikov AM, Kovalenko EI. HLA-DR-Positive NK Cells Expand in Response to Mycobacterium Tuberculosis Antigens and Mediate Mycobacteria-Induced T Cell Activation. Front Immunol 2021; 12:662128. [PMID: 34012446 PMCID: PMC8128146 DOI: 10.3389/fimmu.2021.662128] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/14/2021] [Indexed: 12/27/2022] Open
Abstract
NK cells play an important role in the control of tuberculosis infection: they are not only able to kill the infected cells, but also control the activity of macrophages and development of the adaptive immune response. Still, there is little information on the role of specific NK cell subsets in this network. In this study, we focused on the mycobacteria-driven responses of the NK cells expressing HLA-DR – a type of MHC class II. We have revealed that this subset is increased in the peripheral blood of patients with primary diagnosed tuberculosis, and expands in response to in vitro stimulation with ultrasonically destroyed Mycobacterium tuberculosis cells (sonicate). The expanded HLA-DR+ NK cells had less differentiated phenotype, higher proliferative activity and increased expression of NKp30 and NKp46 receptors. HLA-DR+CD56dim NK cells showed higher IFNγ production and degranulation level than the respective HLA-DR− NK cells in response to both 24 h and 7 day stimulation with sonicate, while HLA-DR+CD56bright NK cells mostly demonstarted similar high responsiveness to the same stimulating conditions as their HLA-DR−CD56bright counterparts. After preliminary incubation with destroyed mycobacteria, cytokine-activated HLA-DR-expressing NK cells were able to mediate mycobacteria-induced and HLA-DR-dependent cytokine production in autologous CD4+ T cells. Thus, functionally active HLA-DR+ cells seem to be one of the NK cell subsets providing an important link to the adaptive immunity.
Collapse
Affiliation(s)
- Sofya A Kust
- Laboratory of Cell Interactions, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Science, Moscow, Russia
| | - Maria A Streltsova
- Laboratory of Cell Interactions, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Science, Moscow, Russia
| | - Alexander V Panteleev
- Laboratory of Biotechnology, Central Tuberculosis Research Institute, Moscow, Russia
| | - Natalya L Karpina
- Diagnostic Outpatient Department, Central Tuberculosis Research Institute, Moscow, Russia
| | - Irina V Lyadova
- Laboratory of Biotechnology, Central Tuberculosis Research Institute, Moscow, Russia.,Laboratory of Cellular and Molecular Basis of Histogenesis, Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| | - Alexander M Sapozhnikov
- Laboratory of Cell Interactions, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Science, Moscow, Russia
| | - Elena I Kovalenko
- Laboratory of Cell Interactions, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Science, Moscow, Russia
| |
Collapse
|
37
|
Rostami MN, Khamesipour A. Potential biomarkers of immune protection in human leishmaniasis. Med Microbiol Immunol 2021; 210:81-100. [PMID: 33934238 PMCID: PMC8088758 DOI: 10.1007/s00430-021-00703-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/22/2021] [Indexed: 12/30/2022]
Abstract
Leishmaniasis is a vector-borne neglected tropical disease endemic in over 100 countries around the world. Available control measures are not always successful, therapeutic options are limited, and there is no vaccine available against human leishmaniasis, although several candidate antigens have been evaluated over the last decades. Plenty of studies have aimed to evaluate the immune response development and a diverse range of host immune factors have been described to be associated with protection or disease progression in leishmaniasis; however, to date, no comprehensive biomarker(s) have been identified as surrogate marker of protection or exacerbation, and lack of enough information remains a barrier for vaccine development. Most of the current understanding of the role of different markers of immune response in leishmaniasis has been collected from experimental animal models. Although the data generated from the animal models are crucial, it might not always be extrapolated to humans. Here, we briefly review the events during Leishmania invasion of host cells and the immune responses induced against Leishmania in animal models and humans and their potential role as a biomarker of protection against human leishmaniasis.
Collapse
Affiliation(s)
| | - Ali Khamesipour
- Center for Research and Training in Skin Diseases and Leprosy, Tehran University of Medical Sciences, 14155-6383, Tehran, Iran.
| |
Collapse
|
38
|
Del Vecchio F, Martinez-Rodriguez V, Schukking M, Cocks A, Broseghini E, Fabbri M. Professional killers: The role of extracellular vesicles in the reciprocal interactions between natural killer, CD8+ cytotoxic T-cells and tumour cells. J Extracell Vesicles 2021; 10:e12075. [PMID: 33815694 PMCID: PMC8015281 DOI: 10.1002/jev2.12075] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 01/27/2021] [Accepted: 02/16/2021] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) mediate the cross‐talk between cancer cells and the cells of the surrounding Tumour Microenvironment (TME). Professional killer cells include Natural Killer (NK) cells and CD8+ Cytotoxic T‐lymphocytes (CTLs), which represent some of the most effective immune defense mechanisms against cancer cells. Recent evidence supports the role of EVs released by NK cells and CTLs in killing cancer cells, paving the road to a possible therapeutic role for such EVs. This review article provides the state‐of‐the‐art knowledge on the role of NK‐ and CTL‐derived EVs as anticancer agents, focusing on the different functions of different sub‐types of EVs. We also reviewed the current knowledge on the effects of cancer‐derived EVs on NK cells and CTLs, identifying areas for future investigation in the emerging new field of EV‐mediated immunotherapy of cancer.
Collapse
Affiliation(s)
- Filippo Del Vecchio
- University of Hawai'i Cancer Center Cancer Biology Program University of Hawai'i at Manoa Honolulu Hawaii USA
| | - Verena Martinez-Rodriguez
- University of Hawai'i Cancer Center Cancer Biology Program University of Hawai'i at Manoa Honolulu Hawaii USA.,Department of Cell and Molecular Biology John A. Burns School of Medicine University of Hawai'i at Manoa Honolulu Hawaii USA
| | - Monique Schukking
- University of Hawai'i Cancer Center Cancer Biology Program University of Hawai'i at Manoa Honolulu Hawaii USA.,Department of Molecular Biosciences & Bioengineering University of Hawai'i at Manoa Honolulu Hawaii USA
| | - Alexander Cocks
- University of Hawai'i Cancer Center Cancer Biology Program University of Hawai'i at Manoa Honolulu Hawaii USA
| | - Elisabetta Broseghini
- University of Hawai'i Cancer Center Cancer Biology Program University of Hawai'i at Manoa Honolulu Hawaii USA.,Department of Experimental, Diagnostic and Specialty Medicine (DIMES) University of Bologna Bologna Italy
| | - Muller Fabbri
- University of Hawai'i Cancer Center Cancer Biology Program University of Hawai'i at Manoa Honolulu Hawaii USA
| |
Collapse
|
39
|
Zeng L, Li C, Jiang H, Chen Y, Li Z, Xu F, Liu R. Total Saponins from Nigella glandulifera Seeds Ameliorate Adjuvant-Induced Rheumatoid Arthritis in Rats by Inhibition of an Inflammatory Response and Bone Erosion. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6613527. [PMID: 33575330 PMCID: PMC7864740 DOI: 10.1155/2021/6613527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/09/2021] [Accepted: 01/13/2021] [Indexed: 12/14/2022]
Abstract
Rheumatoid arthritis (RA) is a widespread inflammatory disease whose clinical manifestations are joint swelling, pain, and disability, affecting approximately 1% of individuals worldwide. Conventional anti-RA drugs currently used in clinic have severe side effects. The present study is aimed at investigating the antiarthritic effects of total saponins from Nigella glandulifera seeds (TSNGS) in rats with adjuvant-induced rheumatoid arthritis (AIA). Arthritis score, paw swelling, and body weight were monitored throughout the period of TSNGS treatment. The histopathological features and levels of cytokines, including IFN-γ, TNF-α, IL-1β, IL-4, IL-6, IL-10, and IL-17A, and OPG/RANKL signaling, were measured to determine the amelioration by TSNGS and its potential mechanisms on the inflammatory response and bone erosion. The differentiation of regulatory T cells (Tregs) in serum was assessed by flow cytometry. The results demonstrate that TSNGS at 10 mg/kg, 50 mg/kg, and 250 mg/kg inhibited AIA-induced clinical score, paw swelling, and histological changes. TSNGS reduced the immune-inflammatory reaction by restoring the secretion and expression of inflammatory cytokines and elevating the proportion of CD4+ CD25+ Tregs, accompanied by an increase in transcription factor Foxp3 levels. TSNGS also displayed bone protection by upregulation of the OPG/RANKL pathway. Collectively, TSNGS inhibited arthritis in AIA rats and so represents a potential novel treatment for RA.
Collapse
Affiliation(s)
- Li Zeng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Chenyang Li
- Key Laboratory of Uighur Medicine of Xinjiang Uygur Autonomous Region, Xinjiang Institute of Materia Medica, Urumqi 830004, China
| | - Hailun Jiang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yan Chen
- Key Laboratory of Uighur Medicine of Xinjiang Uygur Autonomous Region, Xinjiang Institute of Materia Medica, Urumqi 830004, China
| | - Zhuorong Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Fang Xu
- Key Laboratory of Uighur Medicine of Xinjiang Uygur Autonomous Region, Xinjiang Institute of Materia Medica, Urumqi 830004, China
| | - Rui Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
40
|
Campos TM, Novais FO, Saldanha M, Costa R, Lordelo M, Celestino D, Sampaio C, Tavares N, Arruda S, Machado P, Brodskyn C, Scott P, Carvalho EM, Carvalho LP. Granzyme B Produced by Natural Killer Cells Enhances Inflammatory Response and Contributes to the Immunopathology of Cutaneous Leishmaniasis. J Infect Dis 2020; 221:973-982. [PMID: 31748808 DOI: 10.1093/infdis/jiz538] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 10/15/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Skin lesions from patients infected with Leishmania braziliensis has been associated with inflammation induced by cytotoxic CD8+ T cells. In addition, CD8+ T cell-mediated cytotoxicity has not been linked to parasite killing. Meanwhile, the cytotoxic role played by natural killer (NK) cells in cutaneous leishmaniasis (CL) remains poorly understood. METHODS In this study, we observed higher frequencies of NK cells in the peripheral blood of CL patients compared with healthy subjects, and that NK cells expressed more interferon-γ, tumor necrosis factor (TNF), granzyme B, and perforin than CD8+ T cells. RESULTS We also found that most of the cytotoxic activity in CL lesions was triggered by NK cells, and that the high levels of granzyme B produced in CL lesions was associated with larger lesion size. Furthermore, an in vitro blockade of granzyme B was observed to decrease TNF production. CONCCLUSIONS Our data, taken together, suggest an important role by NK cells in inducing inflammation in CL, thereby contributing to disease immunopathology.
Collapse
Affiliation(s)
- Taís M Campos
- Laboratório de Pesquisas Clínicas, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil.,Serviço de Imunologia, Complexo Hospitalar Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Fernanda O Novais
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Maíra Saldanha
- Laboratório Avançado de Saúde Pública, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
| | - Rúbia Costa
- Serviço de Imunologia, Complexo Hospitalar Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Morgana Lordelo
- Laboratório de Interação Parasito-Hospedeiro e Epidemiologia, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
| | - Daniela Celestino
- Serviço de Imunologia, Complexo Hospitalar Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Camilla Sampaio
- Laboratório de Pesquisas Clínicas, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil.,Serviço de Imunologia, Complexo Hospitalar Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Natália Tavares
- Laboratório de Interação Parasito-Hospedeiro e Epidemiologia, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
| | - Sérgio Arruda
- Laboratório Avançado de Saúde Pública, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
| | - Paulo Machado
- Serviço de Imunologia, Complexo Hospitalar Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil.,Instituto Nacional de Ciências e Tecnologia-Doenças Tropicais, Salvador, Brazil
| | - Cláudia Brodskyn
- Laboratório de Interação Parasito-Hospedeiro e Epidemiologia, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
| | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Edgar M Carvalho
- Laboratório de Pesquisas Clínicas, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil.,Serviço de Imunologia, Complexo Hospitalar Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil.,Instituto Nacional de Ciências e Tecnologia-Doenças Tropicais, Salvador, Brazil
| | - Lucas P Carvalho
- Laboratório de Pesquisas Clínicas, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil.,Serviço de Imunologia, Complexo Hospitalar Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil.,Instituto Nacional de Ciências e Tecnologia-Doenças Tropicais, Salvador, Brazil
| |
Collapse
|
41
|
Kim SY, Shin JS, Chung KS, Han HS, Lee HH, Lee JH, Kim SY, Ji YW, Ha Y, Kang J, Rhee YK, Lee KT. Immunostimulatory Effects of Live Lactobacillus sakei K040706 on the CYP-Induced Immunosuppression Mouse Model. Nutrients 2020; 12:nu12113573. [PMID: 33266362 PMCID: PMC7700367 DOI: 10.3390/nu12113573] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/20/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022] Open
Abstract
Our previous studies have shown that heat-killed Lactobacillus sakei K040706 exerts immunostimulatory and anti-inflammatory activities in macrophages, cyclophosphamide (CYP)-treated mice, and dextran sulfate sodium–induced colitis mice. However, the immunostimulatory effects of live Lactobacillus sakei K040706 (live K040706) against CYP-induced immunosuppression and its underlying molecular mechanisms remain unknown. Therefore, we investigated the immunostimulatory effects of live K040706 (108 or 109 colony forming unit (CFU)/day, p.o.) in CYP-induced immunosuppressed mice. Oral administration of live K040706 prevented the CYP-induced decreases in body weight, thymus index, natural killer (NK) cell activity, T and B cell proliferation, and cytokine (interferon (IFN)-γ, interleukin (IL)-2, and IL-12) production. The administration of live K040706 also exerted positive effects on the gut microbiota of CYP-induced mice, resulting in a microbiota composition similar to that of normal mice. Moreover, live K040706 significantly enhanced IL-6 and granulocyte-macrophage colony-stimulating factor (GM-CSF) production in the splenocytes and Peyer’s patch (PP) cells of mice and increased bone marrow (BM) cell proliferation. Taken together, our data indicate that live K040706 may effectively accelerate recovery from CYP-induced immunosuppression, leading to activation of the immune system. Therefore, live K040706 may serve as a potential immunomodulatory agent against immunosuppression.
Collapse
Affiliation(s)
- Seo-Yeon Kim
- Department of Pharmaceutical Biochemistry, College of Pharmacy Kyung Hee University, Seoul 02447, Korea; (S.-Y.K.); (J.-S.S.); (K.-S.C.); (H.-S.H.); (H.-H.L.); (J.-H.L.); (S.-Y.K.)
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Ji-Sun Shin
- Department of Pharmaceutical Biochemistry, College of Pharmacy Kyung Hee University, Seoul 02447, Korea; (S.-Y.K.); (J.-S.S.); (K.-S.C.); (H.-S.H.); (H.-H.L.); (J.-H.L.); (S.-Y.K.)
| | - Kyung-Sook Chung
- Department of Pharmaceutical Biochemistry, College of Pharmacy Kyung Hee University, Seoul 02447, Korea; (S.-Y.K.); (J.-S.S.); (K.-S.C.); (H.-S.H.); (H.-H.L.); (J.-H.L.); (S.-Y.K.)
| | - Hee-Soo Han
- Department of Pharmaceutical Biochemistry, College of Pharmacy Kyung Hee University, Seoul 02447, Korea; (S.-Y.K.); (J.-S.S.); (K.-S.C.); (H.-S.H.); (H.-H.L.); (J.-H.L.); (S.-Y.K.)
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Hwi-Ho Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy Kyung Hee University, Seoul 02447, Korea; (S.-Y.K.); (J.-S.S.); (K.-S.C.); (H.-S.H.); (H.-H.L.); (J.-H.L.); (S.-Y.K.)
| | - Jeong-Hun Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy Kyung Hee University, Seoul 02447, Korea; (S.-Y.K.); (J.-S.S.); (K.-S.C.); (H.-S.H.); (H.-H.L.); (J.-H.L.); (S.-Y.K.)
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Su-Yeon Kim
- Department of Pharmaceutical Biochemistry, College of Pharmacy Kyung Hee University, Seoul 02447, Korea; (S.-Y.K.); (J.-S.S.); (K.-S.C.); (H.-S.H.); (H.-H.L.); (J.-H.L.); (S.-Y.K.)
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Yong Woo Ji
- Department of Ophthalmology, National Health Insurance Service Ilsan Hospital, Goyang 10444, Korea;
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Yejin Ha
- NOVAREX Co. Ltd., 94, Gangni 1-gil, Ochang-eup, Cheongwon-gu, Cheongju-si, Chungcheongbuk-do 363-885, Korea; (Y.H.); (J.K.)
| | - Jooyeon Kang
- NOVAREX Co. Ltd., 94, Gangni 1-gil, Ochang-eup, Cheongwon-gu, Cheongju-si, Chungcheongbuk-do 363-885, Korea; (Y.H.); (J.K.)
| | - Young Kyoung Rhee
- Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Korea;
| | - Kyung-Tae Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy Kyung Hee University, Seoul 02447, Korea; (S.-Y.K.); (J.-S.S.); (K.-S.C.); (H.-S.H.); (H.-H.L.); (J.-H.L.); (S.-Y.K.)
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea
- Correspondence: ; Tel.: +82-2-961-0860; Fax: +82-2-961-0356
| |
Collapse
|
42
|
Ikeogu NM, Akaluka GN, Edechi CA, Salako ES, Onyilagha C, Barazandeh AF, Uzonna JE. Leishmania Immunity: Advancing Immunotherapy and Vaccine Development. Microorganisms 2020; 8:E1201. [PMID: 32784615 PMCID: PMC7465679 DOI: 10.3390/microorganisms8081201] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 01/06/2023] Open
Abstract
Parasitic diseases still constitute a major global health problem affecting billions of people around the world. These diseases are capable of becoming chronic and result in high morbidity and mortality. Worldwide, millions of people die each year from parasitic diseases, with the bulk of those deaths resulting from parasitic protozoan infections. Leishmaniasis, which is a disease caused by over 20 species of the protozoan parasite belonging to the genus Leishmania, is an important neglected disease. According to the World Health Organization (WHO), an estimated 12 million people are currently infected in about 98 countries and about 2 million new cases occur yearly, resulting in about 50,000 deaths each year. Current treatment methods for leishmaniasis are not very effective and often have significant side effects. In this review, we discussed host immunity to leishmaniasis, various treatment options currently being utilized, and the progress of both immunotherapy and vaccine development strategies used so far in leishmaniasis. We concluded with insights into what the future holds toward the fight against this debilitating parasitic disease.
Collapse
Affiliation(s)
- Nnamdi M. Ikeogu
- Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (G.N.A.); (E.S.S.); (C.O.); (A.F.B.)
| | - Gloria N. Akaluka
- Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (G.N.A.); (E.S.S.); (C.O.); (A.F.B.)
| | - Chidalu A. Edechi
- Department of Pathology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P5, Canada;
| | - Enitan S. Salako
- Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (G.N.A.); (E.S.S.); (C.O.); (A.F.B.)
| | - Chukwunonso Onyilagha
- Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (G.N.A.); (E.S.S.); (C.O.); (A.F.B.)
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB R3E 3M4, Canada
| | - Aida F. Barazandeh
- Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (G.N.A.); (E.S.S.); (C.O.); (A.F.B.)
| | - Jude E. Uzonna
- Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (G.N.A.); (E.S.S.); (C.O.); (A.F.B.)
| |
Collapse
|
43
|
Th1 responses in vivo require cell-specific provision of OX40L dictated by environmental cues. Nat Commun 2020; 11:3421. [PMID: 32647184 PMCID: PMC7347572 DOI: 10.1038/s41467-020-17293-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 06/23/2020] [Indexed: 12/24/2022] Open
Abstract
The OX40-OX40L pathway provides crucial co-stimulatory signals for CD4 T cell responses, however the precise cellular interactions critical for OX40L provision in vivo and when these occur, remains unclear. Here, we demonstrate that provision of OX40L by dendritic cells (DCs), but not T cells, B cells nor group 3 innate lymphoid cells (ILC3s), is critical specifically for the effector Th1 response to an acute systemic infection with Listeria monocytogenes (Lm). OX40L expression by DCs is regulated by cross-talk with NK cells, with IFNγ signalling to the DC to enhance OX40L in a mechanism conserved in both mouse and human DCs. Strikingly, DC expression of OX40L is redundant in a chronic intestinal Th1 response and expression by ILC3s is necessary. Collectively these data reveal tissue specific compartmentalisation of the cellular provision of OX40L and define a mechanism controlling DC expression of OX40L in vivo. The OX40-OX40L axis is a crucial component of the costimulatory requirement of CD4 T cell responses. Here, the authors show context and cell type specific expression of OX40L for driving Th1 cell generation during acute and chronic models of infection.
Collapse
|
44
|
Wu W, Metcalf JP. The Role of Type I IFNs in Influenza: Antiviral Superheroes or Immunopathogenic Villains? J Innate Immun 2020; 12:437-447. [PMID: 32564033 PMCID: PMC7747089 DOI: 10.1159/000508379] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/03/2020] [Indexed: 12/29/2022] Open
Abstract
The important role of interferons (IFNs) in antiviral innate immune defense is well established. Although recombinant IFN-α was approved for cancer and chronic viral infection treatment by regulatory agencies in many countries starting in 1986, no IFNs are approved for treatment of influenza A virus (IAV) infection. This is partially due to the complex effects of IFNs in acute influenza infection. IAV attacks the human respiratory system and causes significant morbidity and mortality globally. During influenza infection, depending on the strain of IAV and the individual host, type I IFNs can have protective antiviral effects or can contribute to immunopathology. In the context of virus infection, the immune system has complicated mechanisms regulating the expression and effects of type I IFN to maximize the antiviral response by both activating and enhancing beneficial innate cell function, while limiting immunopathological responses that lead to exaggerated tissue damage. In this review, we summarize the complicated, but important, role of type I IFNs in influenza infections. This includes both protective and harmful effects of these important cytokines during infection.
Collapse
Affiliation(s)
- Wenxin Wu
- Department of Medicine, Pulmonary, Critical Care and Sleep Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA,
| | - Jordan P Metcalf
- Department of Medicine, Pulmonary, Critical Care and Sleep Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Pulmonary Section, Medicine Service, Veterans Affairs Medical Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
45
|
Hashemi E, Malarkannan S. Tissue-Resident NK Cells: Development, Maturation, and Clinical Relevance. Cancers (Basel) 2020; 12:cancers12061553. [PMID: 32545516 PMCID: PMC7352973 DOI: 10.3390/cancers12061553] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/08/2020] [Accepted: 06/08/2020] [Indexed: 12/14/2022] Open
Abstract
Natural killer (NK) cells belong to type 1 innate lymphoid cells (ILC1) and are essential in killing infected or transformed cells. NK cells mediate their effector functions using non-clonotypic germ-line-encoded activation receptors. The utilization of non-polymorphic and conserved activating receptors promoted the conceptual dogma that NK cells are homogeneous with limited but focused immune functions. However, emerging studies reveal that NK cells are highly heterogeneous with divergent immune functions. A distinct combination of several activation and inhibitory receptors form a diverse array of NK cell subsets in both humans and mice. Importantly, one of the central factors that determine NK cell heterogeneity and their divergent functions is their tissue residency. Decades of studies provided strong support that NK cells develop in the bone marrow. However, evolving evidence supports the notion that NK cells also develop and differentiate in tissues. Here, we summarize the molecular basis, phenotypic signatures, and functions of tissue-resident NK cells and compare them with conventional NK cells.
Collapse
Affiliation(s)
- Elaheh Hashemi
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA;
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Subramaniam Malarkannan
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA;
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Correspondence:
| |
Collapse
|
46
|
Tannic acid acts as an agonist of the dopamine D2L receptor, regulates immune responses, and ameliorates experimentally induced colitis in mice. Brain Behav Immun Health 2020; 5:100071. [PMID: 34589853 PMCID: PMC8474654 DOI: 10.1016/j.bbih.2020.100071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 12/13/2022] Open
Abstract
Tannic acid (TA) is an herbal polyphenol containing a galloyl group that has been prescribed to treat gastroenteritis, diarrhea, and irritable bowel syndrome. TA has anti-inflammatory, anti-cancer, and anti-viral properties; however, the molecular mechanisms of these potential therapeutic effects are still largely unknown. Here, we examined the ability of TA to induce anti-inflammatory responses. TA was found to be an agonist of the dopamine D2L receptor. TA reduced interferon (IFN)-γ and interleukin (IL)-1β secretion but upregulated tumor necrosis factor α and IL-10 secretion from lipopolysaccharide (LPS)-stimulated mouse splenocytes. TA also reduced IFN-γ secretion but enhanced IL-10 secretion from anti-cluster of differentiation (CD) 3/CD28 antibody-stimulated splenocytes. An immune subset study confirmed that TA regulated cytokine secretion by various types of immune cells in the context of stimulation with LPS or anti-CD3/CD28 antibodies. Administration of TA to mice with experimentally induced colitis strikingly suppressed weight loss, colon shrinkage, and IL-17 secretion from mesenteric lymph node lymphocytes in response to CD3/CD28 stimulation. These data suggest that TA suppresses inflammatory responses in colitis by regulating cytokine secretion by immune cells in the colon. Tannic acid is an agonist of the dopamine D2L receptor. Tannic acid suppresses IFN-γ secretion by LPS-stimulated splenocytes. Tannic acid modulates anti-CD3/CD28 antibody-stimulated cytokine levels in CD4+ T cells. Tannic acid ameliorates dextran sodium salt (DSS)-induced colitis in C57BL/6 mice. Tannic acid reduces production of IL-17 in DSS-induced colitis.
Collapse
|
47
|
Clarke EC, Bradfute SB. The use of mice lacking type I or both type I and type II interferon responses in research on hemorrhagic fever viruses. Part 1: Potential effects on adaptive immunity and response to vaccination. Antiviral Res 2020; 174:104703. [DOI: 10.1016/j.antiviral.2019.104703] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/10/2019] [Accepted: 12/20/2019] [Indexed: 12/25/2022]
|
48
|
Barsoumian HB, Batra L, Shrestha P, Bowen WS, Zhao H, Egilmez NK, Gomez-Gutierrez JG, Yolcu ES, Shirwan H. A Novel Form of 4-1BBL Prevents Cancer Development via Nonspecific Activation of CD4 + T and Natural Killer Cells. Cancer Res 2019; 79:783-794. [PMID: 30770367 DOI: 10.1158/0008-5472.can-18-2401] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/13/2018] [Accepted: 12/28/2018] [Indexed: 01/11/2023]
Abstract
Costimulation through 4-1BB (CD137) receptor generates robust CD8+ T-effector and memory responses. The only known ligand, 4-1BBL, is a trimeric transmembrane protein that has no costimulatory activity as a soluble molecule. Thus, agonistic antibodies to the receptor have been used for cancer immunotherapy in preclinical models and are currently being evaluated in the clinic. Here, we report that treatment with an oligomeric form of the ligand, SA-4-1BBL, as a single agent is able to protect mice against subsequent tumor challenge irrespective of the tumor type. Protection was long-lasting (>8 weeks) and a bona fide property of SA-4-1BBL, as treatment with an agonistic antibody to the 4-1BB receptor was ineffective in generating immune protection against tumor challenge. Mechanistically, SA-4-1BBL significantly expanded IFNγ-expressing, preexisting memory-like CD44+CD4+ T cells and NK cells in naïve mice as compared with the agonistic antibody. In vivo blockade of IFNγ or depletion of CD4+ T or NK cells, but not CD8+ T or B cells, abrogated the immunopreventive effects of SA-4-1BBL against cancer. SA-4-1BBL as a single agent also exhibited robust efficacy in controlling postsurgical recurrences. This work highlights unexpected features of SA-4-1BBL as a novel immunomodulator with implications for cancer immunoprevention and therapy. SIGNIFICANCE: This study demonstrates the unique and unexpected immunomodulatory features of SA-4-1BBL that bridge innate and adaptive immune responses with both preventive and therapeutic efficacy against cancer.
Collapse
Affiliation(s)
- Hampartsoum B Barsoumian
- Institute for Cellular Therapeutics, University of Louisville, Louisville, Kentucky.,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | - Lalit Batra
- Institute for Cellular Therapeutics, University of Louisville, Louisville, Kentucky.,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | - Pradeep Shrestha
- Institute for Cellular Therapeutics, University of Louisville, Louisville, Kentucky.,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | | | - Hong Zhao
- Institute for Cellular Therapeutics, University of Louisville, Louisville, Kentucky.,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | - Nejat K Egilmez
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | | | - Esma S Yolcu
- Institute for Cellular Therapeutics, University of Louisville, Louisville, Kentucky. .,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky.,FasCure Therapeutics, LLC, Louisville, Kentucky
| | - Haval Shirwan
- Institute for Cellular Therapeutics, University of Louisville, Louisville, Kentucky. .,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| |
Collapse
|
49
|
Cutaneous Leishmaniasis: The Complexity of Host's Effective Immune Response against a Polymorphic Parasitic Disease. J Immunol Res 2019; 2019:2603730. [PMID: 31871953 PMCID: PMC6913332 DOI: 10.1155/2019/2603730] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 10/05/2019] [Accepted: 10/22/2019] [Indexed: 12/14/2022] Open
Abstract
This review is aimed at providing a comprehensive outline of the immune response displayed against cutaneous leishmaniasis (CL), the more common zoonotic infection caused by protozoan parasites of the genus Leishmania. Although of polymorphic clinical presentation, classically CL is characterized by leishmaniotic lesions on the face and extremities of the patients, which can be ulcerative, and even after healing can lead to permanent injuries and disfigurement, affecting significantly their psychological, social, and economic well-being. According a report released by the World Health Organization, the disability-adjusted life years (DALYs) lost due to leishmaniasis are close to 2.4 million, annually there are 1.0–1.5 million new cases of CL, and a numerous population is at risk in the endemic areas. Despite its increasing worldwide incidence, it is one of the so-called neglected tropical diseases. Furthermore, this review provides an overview of the existing knowledge of the host innate and acquired immune response to cutaneous species of Leishmania. The use of animal models and of in vitro studies has improved the understanding of parasite-host interplay and the complexity of immune mechanisms involved. The importance of diagnosis accuracy associated with effective patient management in CL reduction is highlighted. However, the multiple factors involved in CL epizoology associated with the unavailability of vaccines or drugs to prevent infection make difficult to formulate an effective strategy for CL control.
Collapse
|
50
|
Bromodomain inhibitor JQ1 reversibly blocks IFN-γ production. Sci Rep 2019; 9:10280. [PMID: 31311960 PMCID: PMC6635431 DOI: 10.1038/s41598-019-46516-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 06/29/2019] [Indexed: 02/07/2023] Open
Abstract
As a class, ‘BET’ inhibitors disrupt binding of bromodomain and extra-terminal motif (BET) proteins, BRD2, BRD3, BRD4 and BRDT, to acetylated histones preventing recruitment of RNA polymerase 2 to enhancers and promoters, especially super-enhancers, to inhibit gene transcription. As such, BET inhibitors may be useful therapeutics for treatment of cancer and inflammatory disease. For example, the small molecule BET inhibitor, JQ1, selectively represses MYC, an important oncogene regulated by a super-enhancer. IFN-γ, a critical cytokine for both innate and adaptive immune responses, is also regulated by a super-enhancer. Here, we show that JQ1 represses IFN-γ expression in TH1 polarized PBMC cultures, CD4+ memory T cells, and NK cells. JQ1 treatment does not reduce activating chromatin marks at the IFNG locus, but displaces RNA polymerase II from the locus. Further, IFN-γ expression recovers in polarized TH1 cultures following removal of JQ1. Our results show that JQ1 abrogates IFN-γ expression, but repression is reversible. Thus, BET inhibitors may disrupt the normal functions of the innate and adaptive immune response.
Collapse
|