1
|
Liu P, Li G, Zhao N, Liu Q, Liu X, Song X, Shi X, Lun X, Zhang L, Wang J, Lu L. Climate heterogeneity, season variation, and sexual dimorphism modulate the association between MHC II diversity and parasite variation in striped hamster. Integr Zool 2024; 19:1181-1198. [PMID: 38084399 DOI: 10.1111/1749-4877.12791] [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] [Indexed: 11/06/2024]
Abstract
Parasite-mediated selection is widely believed to play a crucial role in maintaining the diversity of the major histocompatibility complex (MHC) genes, which is thought to be maintained through heterozygote advantage, rare-allele advantage, and fluctuating selection. However, the relationship between parasite pressure and MHC diversity has yielded inconsistent findings. These inconsistencies may arise from the influence of environmental factors and individual variations in traits on host-parasite interactions. To address these issues, our study extensively investigated populations of striped hamsters inhabiting regions characterized by environmental heterogeneity. The primary objective was to examine the universality of parasite-mediated selection mechanisms. Our observations revealed the presence of multiple parasite infections, accompanied by spatial and temporal variations in parasite communities and infection patterns among individual hamsters. Specifically, the temperature was found to influence all four parasite indices, while the presence of gamasid mites and parasite richness decreased with increasing precipitation. We also noted significant seasonal variation in parasite dynamics. Moreover, a significant sexual dimorphism was observed with males exhibiting a considerably higher parasite burden compared to their female counterparts. Lastly, we identified the maintenance of MHC polymorphism in striped hamsters as being driven by the heterozygote advantage and fluctuating selection mechanisms. This study underscores the significance of ecological processes in comprehending host-parasite systems and highlights the necessity of considering environmental factors and individual traits when elucidating the mechanisms underlying MHC diversity mediated by parasites.
Collapse
Affiliation(s)
- Pengbo Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Guichang Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ning Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qiyong Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiaobo Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiuping Song
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xinfei Shi
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xinchang Lun
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Lu Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jun Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Liang Lu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
2
|
Teixeira TL, Teixeira SC, Borges BC, Servato JPS, Oliveira ECMD, Velikkakam T, Silva CVD. Trypanosoma cruzi P21 protein exacerbates Leishmania (L.) amazonensis infection. Exp Parasitol 2024; 267:108856. [PMID: 39461608 DOI: 10.1016/j.exppara.2024.108856] [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: 03/26/2024] [Revised: 09/26/2024] [Accepted: 10/24/2024] [Indexed: 10/29/2024]
Abstract
The protozoan parasite Trypanosoma cruzi, the etiological agent of Chagas disease, affects millions of people worldwide. Current treatments rely on drugs effective only in the acute phase, making the search for new therapeutic targets a priority. While a recombinant protein based on T. cruzi P21 (rP21) exhibits immunomodulatory properties and contributes to controlling parasitism and inflammation during T. cruzi infection, its efficacy against other trypanosomatids remains unexplored. This study investigated the impact of rP21 on Leishmania (L.) amazonensis infection in a murine model. Contrary to our expectations, treatment with rP21 did not ameliorate L. (L.) amazonensis infection. Instead, rP21 treatment resulted in increased parasite load in the paws of infected BALB/c mice, evidenced by larger lesion sizes and higher parasite burdens, accompanied by an intensified inflammatory infiltrate in the paw tissue. These findings suggest that despite its promising effects in the context of T. cruzi infection, rP21 may not be a suitable therapeutic candidate for L. amazonensis infection and might even exacerbate disease.
Collapse
Affiliation(s)
- Thaise Lara Teixeira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Samuel Cota Teixeira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Bruna Cristina Borges
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | | | | | - Teresiama Velikkakam
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Claudio Vieira da Silva
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil.
| |
Collapse
|
3
|
Ihedioha OC, Marcarian HQ, Sivakoses A, Beverley SM, McMahon-Pratt D, Bothwell ALM. Leishmania major surface components and DKK1 signalling via LRP6 promote migration and longevity of neutrophils in the infection site. Front Immunol 2024; 15:1473133. [PMID: 39502693 PMCID: PMC11534728 DOI: 10.3389/fimmu.2024.1473133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/30/2024] [Indexed: 11/08/2024] Open
Abstract
Background Host-related factors highly regulate the increased circulation of neutrophils during Leishmania infection. Platelet-derived Dickkopf-1 (DKK1) is established as a high-affinity ligand to LRP6. Recently, we demonstrated that DKK1 upregulates leukocyte-platelet aggregation, infiltration of neutrophils to the draining lymph node and Th2 differentiation during Leishmania infection, suggesting the potential involvement of the DKK1-LRP6 signalling pathway in neutrophil migration in infectious diseases. Results In this study, we further explored the potential role of DKK1-LRP6 signalling in the migration and longevity of activated neutrophils in the infection site using BALB/c mice with PMNs deficient in LRP6 (LRP6NKO) or BALB/c mice deficient in both PMN LRP6 and platelet DKK1 (LRP6NKO DKK1PKO). Relative to the infected wild-type BALB/c mice, reduced neutrophil activation at the infection site of LRP6NKO or LRP6NKO DKK1PKO mice was noted. The neutrophils obtained from either infected LRP6NKO or LRP6NKO DKK1PKO mice additionally showed a high level of apoptosis. Notably, the level of LRP6 expressing neutrophils was elevated in infected BALB/c mice. Relative to infected BALB/c mice, a significant reduction in parasite load was observed in both LRP6NKO and LRP6NKO DKK1PKO infected mice. Notably, DKK1 levels were comparable in the LRP6NKO and BALB/c mice in response to infection, indicating that PMN activation is the major pathway for DKK1 in promoting parasitemia. Parasite-specific components also play a crucial role in modulating neutrophil circulation in Leishmania disease. Thus, we further determine the contribution of Leishmania membrane components in the migration of neutrophils to the infection site using null mutants deficient in LPG synthesis (Δlpg1- ) or lacking all ether phospholipids (plasmalogens, LPG, and GIPLs) synthesis (Δads1- ). Relative to the WT controls, Δads1- parasite-infected mice showed a sustained decrease in neutrophils and neutrophil-platelet aggregates (for at least 14 days PI), while neutrophils returned to normal in Δlpg1- parasite-infected mice after day 3 PI. Conclusion Our results suggest that DKK1 signalling and Leishmania pathogen-associated molecular patterns appear to regulate the migration and sustenance of viable activated neutrophils in the infection site resulting in chronic type 2 cell-mediated inflammation.
Collapse
Affiliation(s)
- Olivia C. Ihedioha
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Haley Q. Marcarian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Anutr Sivakoses
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Stephen M. Beverley
- Department of Molecular Microbiology, Washington University School of Medicine in St Louis, St. Louis, MO, United States
| | - Diane McMahon-Pratt
- Department of Epidemiology of Infectious Diseases, Yale School of Public Health, New Haven, CT, United States
| | - Alfred L. M. Bothwell
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
4
|
DeSouza-Vieira T, Pretti MAM, Lima Gomes PS, Paula-Neto HA, Goundry A, Nascimento MT, Ganesan S, Gonçalves da Silva T, Kamenyeva O, Kabat J, Manzella-Lapeira J, B. Canto F, Fraga-Junior VDS, Eustáquio Lopes M, Gomes Vaz L, Pessenda G, Paun A, Freitas-Mesquita AL, Meyer-Fernandes JR, Boroni M, Bellio M, Batista Menezes G, Brzostowski J, Mottram J, Sacks D, Lima APCA, Saraiva EM. Functional plasticity shapes neutrophil response to Leishmania major infection in susceptible and resistant strains of mice. PLoS Pathog 2024; 20:e1012592. [PMID: 39378227 PMCID: PMC11488723 DOI: 10.1371/journal.ppat.1012592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/18/2024] [Accepted: 09/14/2024] [Indexed: 10/10/2024] Open
Abstract
Neutrophils rapidly infiltrate sites of infection and possess several microbicidal strategies, such as neutrophil extracellular traps release and phagocytosis. Enhanced neutrophil infiltration is associated with higher susceptibility to Leishmania infection, but neutrophil effector response contribution to this phenotype is uncertain. Here, we show that neutrophils from susceptible BALB/c mice (B/c) produce more NETs in response to Leishmania major than those from resistant C57BL/6 mice (B6), which are more phagocytic. The absence of neutrophil elastase contributes to phagocytosis regulation. Microarray analysis shows enrichment of genes involved in NET formation (mpo, pi3kcg, il1b) in B/c, while B6 shows upregulation of genes involved in phagocytosis and cell death (Arhgap12, casp9, mlkl, FasL). scRNA-seq in L. major-infected B6 showed heterogeneity in the pool of intralesional neutrophils, and we identified the N1 subset as the putative subpopulation involved with phagocytosis. In vivo, imaging validates NET formation in infected B/c ears where NETing neutrophils were mainly uninfected cells. NET digestion in vivo augmented parasite lymphatic drainage. Hence, a balance between NET formation and phagocytosis in neutrophils may contribute to the divergent phenotype observed in these mice.
Collapse
Affiliation(s)
- Thiago DeSouza-Vieira
- Laboratório de Imunobiologia das Leishmanioses, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Marco Antônio M. Pretti
- Laboratório de Bioinformática e Biologia Computacional, Divisão de Pesquisa Experimental Translacional, Instituto Nacional do Câncer (INCA), Rio de Janeiro, Brasil
| | - Phillipe Souza Lima Gomes
- Laboratório de Imunobiologia das Leishmanioses, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Heitor A. Paula-Neto
- Laboratório de Alvos Moleculares, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Amy Goundry
- Laboratório de Bioquímica e Biologia Molecular de Proteases, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Michelle T. Nascimento
- Laboratório de Imunobiologia das Leishmanioses, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Sundar Ganesan
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Triciana Gonçalves da Silva
- National Center for Structural Biology and Bioimaging, CENABIO, Universidade Federal do Rio de Janeiro, Brazil
| | - Olena Kamenyeva
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Juraj Kabat
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Javier Manzella-Lapeira
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Fábio B. Canto
- Laboratório de Tolerância Imunológica e Homeostase Linfocitária, Departamento de Imunobiologia, Universidade Federal Fluminense, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Vanderlei da Silva Fraga-Junior
- Laboratório de Imunologia Molecular e Celular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Mateus Eustáquio Lopes
- Centro de Biologia Gastrointestinal, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Leonardo Gomes Vaz
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Pampulha, Belo Horizonte, Minas Gerais, Brasil
| | - Gabriela Pessenda
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Andrea Paun
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Anita L. Freitas-Mesquita
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - José Roberto Meyer-Fernandes
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Mariana Boroni
- Laboratório de Bioinformática e Biologia Computacional, Divisão de Pesquisa Experimental Translacional, Instituto Nacional do Câncer (INCA), Rio de Janeiro, Brasil
| | - Maria Bellio
- Laboratório de Imunobiologia, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Gustavo Batista Menezes
- Centro de Biologia Gastrointestinal, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Joseph Brzostowski
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Jeremy Mottram
- York Biomedical Research Institute and Department of Biology, University of York, York, United Kingdom
| | - David Sacks
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ana Paula C. A. Lima
- Laboratório de Bioquímica e Biologia Molecular de Proteases, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Elvira M. Saraiva
- Laboratório de Imunobiologia das Leishmanioses, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| |
Collapse
|
5
|
Paul R, Chakrabarty A, Samanta S, Dey S, Pandey R, Maji S, Pezacki AT, Chang CJ, Datta R, Gupta A. Leishmania major-induced alteration of host cellular and systemic copper homeostasis drives the fate of infection. Commun Biol 2024; 7:1226. [PMID: 39349621 PMCID: PMC11442737 DOI: 10.1038/s42003-024-06716-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 08/12/2024] [Indexed: 10/04/2024] Open
Abstract
Copper plays a key role in host-pathogen interaction. We find that during Leishmania major infection, the parasite-harboring macrophage regulates its copper homeostasis pathway in a way to facilitate copper-mediated neutralization of the pathogen. Copper-ATPase ATP7A transports copper to amastigote-harboring phagolysosomes to induce stress on parasites. Leishmania in order to evade the copper stress, utilizes a variety of manipulative measures to lower the host-induced copper stress. It induces deglycosylation and degradation of host-ATP7A and downregulation of copper importer, CTR1 by cysteine oxidation. Additionally, Leishmania induces CTR1 endocytosis that arrests copper uptake. In mouse model of infection, we report an increase in systemic bioavailable copper in infected animals. Heart acts as the major organ for diverting its copper reserves to systemic circulation to fight-off infection by downregulating its CTR1. Our study explores reciprocal mechanism of manipulation of host copper homeostasis pathway by macrophage and Leishmania to gain respective advantages in host-pathogen interaction.
Collapse
Affiliation(s)
- Rupam Paul
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India.
| | - Adrija Chakrabarty
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Suman Samanta
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Swastika Dey
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Raviranjan Pandey
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Saptarshi Maji
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Aidan T Pezacki
- Department of Chemistry, University of California Berkeley, Berkeley, CA, USA
| | - Christopher J Chang
- Department of Chemistry, University of California Berkeley, Berkeley, CA, USA
| | - Rupak Datta
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Arnab Gupta
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India.
| |
Collapse
|
6
|
Mihlan M, Wissmann S, Gavrilov A, Kaltenbach L, Britz M, Franke K, Hummel B, Imle A, Suzuki R, Stecher M, Glaser KM, Lorentz A, Carmeliet P, Yokomizo T, Hilgendorf I, Sawarkar R, Diz-Muñoz A, Buescher JM, Mittler G, Maurer M, Krause K, Babina M, Erpenbeck L, Frank M, Rambold AS, Lämmermann T. Neutrophil trapping and nexocytosis, mast cell-mediated processes for inflammatory signal relay. Cell 2024; 187:5316-5335.e28. [PMID: 39096902 DOI: 10.1016/j.cell.2024.07.014] [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: 07/31/2023] [Revised: 04/10/2024] [Accepted: 07/08/2024] [Indexed: 08/05/2024]
Abstract
Neutrophils are sentinel immune cells with essential roles for antimicrobial defense. Most of our knowledge on neutrophil tissue navigation derived from wounding and infection models, whereas allergic conditions remained largely neglected. Here, we analyzed allergen-challenged mouse tissues and discovered that degranulating mast cells (MCs) trap living neutrophils inside them. MCs release the attractant leukotriene B4 to re-route neutrophils toward them, thus exploiting a chemotactic system that neutrophils normally use for intercellular communication. After MC intracellular trap (MIT) formation, neutrophils die, but their undigested material remains inside MC vacuoles over days. MCs benefit from MIT formation, increasing their functional and metabolic fitness. Additionally, they are more pro-inflammatory and can exocytose active neutrophilic compounds with a time delay (nexocytosis), eliciting a type 1 interferon response in surrounding macrophages. Together, our study highlights neutrophil trapping and nexocytosis as MC-mediated processes, which may relay neutrophilic features over the course of chronic allergic inflammation.
Collapse
Affiliation(s)
- Michael Mihlan
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Münster 48149, Germany.
| | - Stefanie Wissmann
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Institute for Biomechanics, ETH Zürich, Zürich 8092, Switzerland
| | - Alina Gavrilov
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Roche Pharma Research and Early Development (pRED), Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology (CMI2O), Roche Innovation Center, Basel 4070, Switzerland
| | - Lukas Kaltenbach
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Marie Britz
- Department of Dermatology, Universitätsklinikum Münster, Münster 48149, Germany
| | - Kristin Franke
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Berlin 12203, Germany; Charité-Universitätsmedizin Berlin, Institute of Allergology, Berlin 12203, Germany
| | - Barbara Hummel
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Andrea Imle
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg 69117, Germany
| | - Ryo Suzuki
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Manuel Stecher
- Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Münster 48149, Germany
| | - Katharina M Glaser
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Institut Curie, PSL Research University, INSERM U932, Paris 75005, France
| | - Axel Lorentz
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart 70593, Germany
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven 3000, Belgium; Center for Biotechnology, Khalifa University of Science and Technology, PO Box 127788, Abu Dhabi, United Arab Emirates
| | - Takehiko Yokomizo
- Department of Biochemistry, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology, University Heart Center and Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Ritwick Sawarkar
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Medical Research Council (MRC) Toxicology Unit and Department of Genetics, University of Cambridge, Cambridge CB21QR, UK
| | - Alba Diz-Muñoz
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg 69117, Germany
| | - Joerg M Buescher
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Gerhard Mittler
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Marcus Maurer
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Berlin 12203, Germany; Charité-Universitätsmedizin Berlin, Institute of Allergology, Berlin 12203, Germany
| | - Karoline Krause
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Berlin 12203, Germany; Charité-Universitätsmedizin Berlin, Institute of Allergology, Berlin 12203, Germany
| | - Magda Babina
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Berlin 12203, Germany; Charité-Universitätsmedizin Berlin, Institute of Allergology, Berlin 12203, Germany
| | - Luise Erpenbeck
- Department of Dermatology, Universitätsklinikum Münster, Münster 48149, Germany
| | - Marcus Frank
- Medical Biology and Electron Microscopy Center, Rostock University Medical Center, Rostock 18057, Germany; Department Life, Light and Matter, Rostock University, Rostock 18051, Germany
| | - Angelika S Rambold
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Tim Lämmermann
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Münster 48149, Germany.
| |
Collapse
|
7
|
Goris M, Passelli K, Peyvandi S, Díaz-Varela M, Billion O, Prat-Luri B, Demarco B, Desponds C, Termote M, Iniguez E, Dey S, Malissen B, Kamhawi S, Hurrell BP, Broz P, Tacchini-Cottier F. NLRP1-dependent activation of Gasdermin D in neutrophils controls cutaneous leishmaniasis. PLoS Pathog 2024; 20:e1012527. [PMID: 39250503 PMCID: PMC11412672 DOI: 10.1371/journal.ppat.1012527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 09/19/2024] [Accepted: 08/22/2024] [Indexed: 09/11/2024] Open
Abstract
Intracellular pathogens that replicate in host myeloid cells have devised ways to inhibit the cell's killing machinery. Pyroptosis is one of the host strategies used to reduce the pathogen replicating niche and thereby control its expansion. The intracellular Leishmania parasites can survive and use neutrophils as a silent entry niche, favoring subsequent parasite dissemination into the host. Here, we show that Leishmania mexicana induces NLRP1- and caspase-1-dependent Gasdermin D (GSDMD)-mediated pyroptosis in neutrophils, a process critical to control the parasite-induced pathology. In the absence of GSDMD, we observe an increased number of infected dermal neutrophils two days post-infection. Using adoptive neutrophil transfer in neutropenic mice, we show that pyroptosis contributes to the regulation of the neutrophil niche early after infection. The critical role of neutrophil pyroptosis and its positive influence on the regulation of the disease outcome was further demonstrated following infection of mice with neutrophil-specific deletion of GSDMD. Thus, our study establishes neutrophil pyroptosis as a critical regulator of leishmaniasis pathology.
Collapse
Affiliation(s)
- Michiel Goris
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Katiuska Passelli
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Sanam Peyvandi
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Miriam Díaz-Varela
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Oaklyne Billion
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Borja Prat-Luri
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Benjamin Demarco
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Chantal Desponds
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Manon Termote
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Eva Iniguez
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Somaditya Dey
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
- Post Graduate Department of Zoology, Barasat Government College, Barasat, West Bengal, India
| | - Bernard Malissen
- INSERM, CNRS, Centre D’Immunologie de Marseille-Luminy, Aix-Marseille Université, Marseille, France
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Benjamin P. Hurrell
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Petr Broz
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Fabienne Tacchini-Cottier
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
8
|
Martins VD, Vaz L, Barbosa SC, Paixão PHDM, Torres L, de Oliveira MFA, Oliveira MDA, Vieira LQ, de Faria AMC, Maioli TU. Obesity alters the macrophages' response to Leishmania major in C57BL/6 mice. J Leukoc Biol 2024:qiae171. [PMID: 39213305 DOI: 10.1093/jleuko/qiae171] [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: 11/02/2023] [Revised: 06/12/2024] [Indexed: 09/04/2024] Open
Abstract
Obesity is a global pandemic associated with several comorbidities, such as cardiovascular diseases and type 2 diabetes. It is also a predisposing factor for infectious diseases, increasing mortality rates. Moreover, diet-induced obesity can cause metabolic fluctuations that affect macrophage differentiation in various organs. In this sense, we investigated how bone marrow-derived macrophages and tissue-resident macrophages in the skin, which have been differentiated in a host with metabolic syndrome and with previous inflammatory burden, respond to Leishmania major infection. Our findings suggest that bone marrow-derived macrophages from obese C57BL/6 mice, even when cultivated in vitro with inflammatory stimuli, are more susceptible to L. major. These macrophages produce less tumor necrosing factor (TNF) and nitric oxide (NO) and show higher arginase activity. Furthermore, obese mice infected with an intermediate dose of L. major in the skin had more severe lesions when analyzed for ulceration, diameter, thickness, and parasite burden. The increase in lesion severity in obese mice was associated with a higher frequency of tissue-resident macrophages, which are less efficient in killing parasites. We also used CCR2-/- mice, which predominantly have tissue-resident macrophages, and found that lesion resolution was delayed in association with CCR2 deficiency. Additionally, obesity potentiated tissue damage, resulting in higher frequency of tissue-resident macrophages. Our results demonstrate that obesity can alter macrophage responses to infection, leading to increased susceptibility to L. major and more severe cutaneous leishmaniasis. These findings may have important implications for managing obesity-related infections and the development of new therapies for cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Vinicius Dantas Martins
- Postgraduate Program in Biochemistry and Immunology, Biological Sciences Institution, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, CEP 31270-672, Belo Horizonte, Brazil
| | - Leonardo Vaz
- Postgraduate Program in Biochemistry and Immunology, Biological Sciences Institution, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, CEP 31270-672, Belo Horizonte, Brazil
| | - Sara Candida Barbosa
- Postgraduate Program in Biochemistry and Immunology, Biological Sciences Institution, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, CEP 31270-672, Belo Horizonte, Brazil
| | - Pierre Henrique de Menezes Paixão
- Postgraduate Program in Pathology, Biological Sciences Institution, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, CEP 31270-672, Belo Horizonte, Brazil
| | - Licia Torres
- Postgraduate Program in Biochemistry and Immunology, Biological Sciences Institution, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, CEP 31270-672, Belo Horizonte, Brazil
- Postgraduate Program in Nutrition and Health, School of Nursing, Universidade Federal de Minas Gerais, Avenida Alfredo Balena, 190, CEP 30130-100, Belo Horizonte, Brazil
| | - Marcos Felipe Andrade de Oliveira
- Postgraduate Program in Biochemistry and Immunology, Biological Sciences Institution, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, CEP 31270-672, Belo Horizonte, Brazil
| | - Mariana de Almeida Oliveira
- Postgraduate Program in Biochemistry and Immunology, Biological Sciences Institution, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, CEP 31270-672, Belo Horizonte, Brazil
| | - Leda Quercia Vieira
- Postgraduate Program in Biochemistry and Immunology, Biological Sciences Institution, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, CEP 31270-672, Belo Horizonte, Brazil
| | - Ana Maria Caetano de Faria
- Postgraduate Program in Biochemistry and Immunology, Biological Sciences Institution, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, CEP 31270-672, Belo Horizonte, Brazil
| | - Tatiani Uceli Maioli
- Postgraduate Program in Biochemistry and Immunology, Biological Sciences Institution, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, CEP 31270-672, Belo Horizonte, Brazil
- Postgraduate Program in Nutrition and Health, School of Nursing, Universidade Federal de Minas Gerais, Avenida Alfredo Balena, 190, CEP 30130-100, Belo Horizonte, Brazil
| |
Collapse
|
9
|
Xu C, Nedergaard M, Fowell DJ, Friedl P, Ji N. Multiphoton fluorescence microscopy for in vivo imaging. Cell 2024; 187:4458-4487. [PMID: 39178829 PMCID: PMC11373887 DOI: 10.1016/j.cell.2024.07.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/26/2024]
Abstract
Multiphoton fluorescence microscopy (MPFM) has been a game-changer for optical imaging, particularly for studying biological tissues deep within living organisms. MPFM overcomes the strong scattering of light in heterogeneous tissue by utilizing nonlinear excitation that confines fluorescence emission mostly to the microscope focal volume. This enables high-resolution imaging deep within intact tissue and has opened new avenues for structural and functional studies. MPFM has found widespread applications and has led to numerous scientific discoveries and insights into complex biological processes. Today, MPFM is an indispensable tool in many research communities. Its versatility and effectiveness make it a go-to technique for researchers investigating biological phenomena at the cellular and subcellular levels in their native environments. In this Review, the principles, implementations, capabilities, and limitations of MPFM are presented. Three application areas of MPFM, neuroscience, cancer biology, and immunology, are reviewed in detail and serve as examples for applying MPFM to biological research.
Collapse
Affiliation(s)
- Chris Xu
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY 14850, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Nørre Alle 3B, 2200 Copenhagen, Denmark; University of Rochester Medical School, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Deborah J Fowell
- Department of Microbiology & Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Peter Friedl
- Department of Medical BioSciences, Radboud University Medical Centre, Geert Grooteplein 26-28, Nijmegen HB 6500, the Netherlands
| | - Na Ji
- Department of Neuroscience, Department of Physics, University of California Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
10
|
Borges-Fernandes LO, de Lima Moreira M, Pereira VHS, Pascoal-Xavier MA, Lopes Ribeiro Á, da Costa-Rocha IA, Lopes LR, Moreira GTC, Araújo MSDS, Teixeira-Carvalho A, Brito-de-Sousa JP, de Carvalho AL, Mourão MVA, Campos FA, Borges M, Carneiro M, Tsuji M, Martins-Filho OA, Coelho-dos-Reis JGA, Peruhype-Magalhães V. MR1 blockade drives differential impact on integrative signatures based on circuits of circulating immune cells and soluble mediators in visceral leishmaniasis. Front Immunol 2024; 15:1373498. [PMID: 39192975 PMCID: PMC11347828 DOI: 10.3389/fimmu.2024.1373498] [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: 01/19/2024] [Accepted: 07/15/2024] [Indexed: 08/29/2024] Open
Abstract
Introduction Visceral leishmaniasis (VL) is an important tropical and neglected disease and represents a serious global health problem. The initial interaction between the phagocytes and the parasite is crucial to determine the pathogen's capacity to initiate infection and it shapes the subsequent immune response that will develop. While type-1 T-cells induce IL-6, IL-1β, TNF-α, and IL-12 production by monocytes/macrophages to fight the infection, type-2 T-cells are associated with a regulatory phenotype (IL-10 and TGF-β) and successful infection establishment. Recently, our group demonstrated the role of an important Th1/Th17 T-cell population, the mucosal-associated invariant T (MAIT) cells, in VL. MAIT cells can respond to L. infantum by producing TNF-α and IFN-γ upon MR1-dependent activation. Objective and methods Here, we describe the impact of the MR1-blockage on L. infantum internalization on the functional profile of circulating neutrophils and monocytes as well as the impact of the MR1-blockage on the soluble mediator signatures of in vitro whole blood cultures. Results Overall, our data showed that VL patients presents higher percentage of activated neutrophils than asymptomatic and non-infected controls. In addition, MR1 blockade led to lower TNF-α and TGF-β production by non-activated neutrophils from asymptomatic individuals. Moreover, TNF-α and IL-10 production by monocytes was higher in VL patients. In the analysis of soluble mediators produced in vitro, MR1-blockade induced a decrease of IFN-γ and an increase of IL-10, IL-27 and IL-33 in the cell cultures of AS group, a cytokine pattern associated with type 2 deleterious response. Discussion and conclusion These data corroborate the hypothesis that MR1-restricted responses are associated to a protective role during Leishmania infection.
Collapse
Affiliation(s)
| | - Marcela de Lima Moreira
- René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ-MINAS), Belo Horizonte, Minas Gerais, Brazil
| | | | - Marcelo Antônio Pascoal-Xavier
- René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ-MINAS), Belo Horizonte, Minas Gerais, Brazil
- School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ágata Lopes Ribeiro
- René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ-MINAS), Belo Horizonte, Minas Gerais, Brazil
- Basic and Applied Virology Laboratory, Department of Microbiology, Institute for Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Ludmila Rosa Lopes
- René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ-MINAS), Belo Horizonte, Minas Gerais, Brazil
| | | | | | - Andréa Teixeira-Carvalho
- René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ-MINAS), Belo Horizonte, Minas Gerais, Brazil
| | | | - Andrea Lucchesi de Carvalho
- João Paulo II Children’s Hospital, Fundação Hospitalar do Estado de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Flávia Alves Campos
- João Paulo II Children’s Hospital, Fundação Hospitalar do Estado de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marineide Borges
- João Paulo II Children’s Hospital, Fundação Hospitalar do Estado de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mariângela Carneiro
- Parasitology Department, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Moriya Tsuji
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
- Division of Infectious Disease, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | | | - Jordana Grazziela Alves Coelho-dos-Reis
- René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ-MINAS), Belo Horizonte, Minas Gerais, Brazil
- Basic and Applied Virology Laboratory, Department of Microbiology, Institute for Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | |
Collapse
|
11
|
Zhou Q, Hu Y, You Y, Gao Y, Wang X, Qin L. Functional analysis of OmpA and its contribution to pathogenesis of Edwardsiella tarda. Microb Pathog 2024; 193:106760. [PMID: 38914348 DOI: 10.1016/j.micpath.2024.106760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 03/27/2024] [Accepted: 06/21/2024] [Indexed: 06/26/2024]
Abstract
Outer membrane protein A (OmpA), a major component of outer membrane proteins in gram-negative bacteria, is considered to be an important virulence factor in various pathogenic bacteria, but its underlying mechanisms involved in pathogenic process of Edwardsiella tarda has not yet been fully elucidated. E. tarda is an important facultative intracellular pathogen with a broad host range. This bacterium could survive and replicate in macrophages as an escape mechanism from the host defense. To address the functions of OmpA and its potential roles in the pathogenesis of E. tarda, ΔompA mutant strain and ΔompA-C complementary strain were constructed by the allelic exchange method in this study. Here, we demonstrate that the abilities of motility, biofilm formation and adherence to RAW264.7 cells of ΔompA were significantly impaired, although there was no difference in growth between wild-type (WT) strain and ΔompA. Moreover, inactivation of ompA rendered E. tarda more sensitive to oxidative, heat shock and osmotic stress, which simulate the in vivo conditions that E. tarda encounters within the intramacrophage environment. Consist with this observation, ΔompA was also found to be markedly attenuated for growth within macrophages. In addition, compared with the WT strain, ΔompA activated macrophages to release more inflammatory mediators, including tumor necrosis factor alpha (TNF-α), reactive oxygen species (ROS) and nitric oxide (NO). However, flow cytometry analysis revealed that ΔompA induced less apoptosis of RAW264.7 cells as compared with WT strain, characterized by decreased Annexin V binding and the activation of caspase-3. Overall, our findings suggest an importance of OmpA to E. tarda and provide the first comprehensive insight into its functions and potential roles in the pathogenesis of E. tarda, including its effect on interaction with macrophages.
Collapse
Affiliation(s)
- Quan Zhou
- School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang, China
| | - Yushuai Hu
- School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang, China
| | - Yicheng You
- School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang, China
| | - Yingli Gao
- School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang, China
| | - Xingqiang Wang
- School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang, China
| | - Lei Qin
- School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang, China.
| |
Collapse
|
12
|
Scaramele NF, Troiano JA, Felix JDS, Costa SF, Almeida MC, Florencio de Athayde FR, Soares MF, Lopes MFDS, Furlan ADO, de Lima VMF, Lopes FL. Leishmania infantum infection modulates messenger RNA, microRNA and long non-coding RNA expression in human neutrophils in vitro. PLoS Negl Trop Dis 2024; 18:e0012318. [PMID: 39028711 PMCID: PMC11259272 DOI: 10.1371/journal.pntd.0012318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/25/2024] [Indexed: 07/21/2024] Open
Abstract
In the Americas, L. infantum (syn. chagasi) is the main cause of human visceral leishmaniasis. The role of neutrophils as part of the innate response to Leishmania spp. infection is dubious and varies according to the species causing the infection. Global expression of coding RNAs, microRNAs and long non-coding RNAs changes as part of the immune response against pathogens. Changes in mRNA and non-coding RNA expression resulting from infection by Leishmania spp. are widely studied in macrophages, but scarce in neutrophils, the first cell to encounter the trypanosomatid, especially following infection by L. infantum. Herein, we aimed to understand the expression patterns of coding and non-coding transcripts during acute in vitro infection of human neutrophils by L. infantum. We isolated neutrophils from whole blood of healthy male donors (n = 5) and split into groups: 1) infected with L. infantum (MOI = 5:1), and 2) uninfected controls. After 3 hours of exposure of infected group to promastigotes of L. infantum, followed by 17 hours of incubation, total RNA was extracted and total RNA-Seq and miRNA microarray were performed. A total of 212 genes were differentially expressed in neutrophils following RNA-Seq analysis (log2(FC)±0.58, FDR≤0.05). In vitro infection with L. infantum upregulated the expression of 197 and reduced the expression of 92 miRNAs in human neutrophils (FC±2, FDR≤0.01). Lastly, 5 downregulated genes were classified as lncRNA, and of the 10 upregulated genes, there was only 1 lncRNA. Further bioinformatic analysis indicated that changes in the transcriptome and microtranscriptome of neutrophils, following in vitro infection with L. infantum, may impair phagocytosis, apoptosis and decrease nitric oxide production. Our work sheds light on several mechanisms used by L. infantum to control neutrophil-mediated immune response and identifies several targets for future functional studies, aiming at the development of preventive or curative treatments for this prevalent zoonosis.
Collapse
Affiliation(s)
- Natália Francisco Scaramele
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Jéssica Antonini Troiano
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Juliana de Souza Felix
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Sidnei Ferro Costa
- Department of Animal Clinic, Surgery and Reproduction, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Mariana Cordeiro Almeida
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Flávia Regina Florencio de Athayde
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Matheus Fujimura Soares
- Department of Animal Clinic, Surgery and Reproduction, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Maria Fernanda da Silva Lopes
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Amanda de Oliveira Furlan
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Valéria Marçal Felix de Lima
- Department of Animal Clinic, Surgery and Reproduction, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Flavia Lombardi Lopes
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| |
Collapse
|
13
|
Fowler EA, Farias Amorim C, Mostacada K, Yan A, Amorim Sacramento L, Stanco RA, Hales ED, Varkey A, Zong W, Wu GD, de Oliveira CI, Collins PL, Novais FO. Neutrophil-mediated hypoxia drives pathogenic CD8+ T cell responses in cutaneous leishmaniasis. J Clin Invest 2024; 134:e177992. [PMID: 38833303 PMCID: PMC11245163 DOI: 10.1172/jci177992] [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: 11/27/2023] [Accepted: 05/17/2024] [Indexed: 06/06/2024] Open
Abstract
Cutaneous leishmaniasis caused by Leishmania parasites exhibits a wide range of clinical manifestations. Although parasites influence disease severity, cytolytic CD8+ T cell responses mediate disease. Although these responses originate in the lymph node, we found that expression of the cytolytic effector molecule granzyme B was restricted to lesional CD8+ T cells in Leishmania-infected mice, suggesting that local cues within inflamed skin induced cytolytic function. Expression of Blimp-1 (Prdm1), a transcription factor necessary for cytolytic CD8+ T cell differentiation, was driven by hypoxia within the inflamed skin. Hypoxia was further enhanced by the recruitment of neutrophils that consumed oxygen to produce ROS and ultimately increased the hypoxic state and granzyme B expression in CD8+ T cells. Importantly, lesions from patients with cutaneous leishmaniasis exhibited hypoxia transcription signatures that correlated with the presence of neutrophils. Thus, targeting hypoxia-driven signals that support local differentiation of cytolytic CD8+ T cells may improve the prognosis for patients with cutaneous leishmaniasis, as well as for other inflammatory skin diseases in which cytolytic CD8+ T cells contribute to pathogenesis.
Collapse
Affiliation(s)
- Erin A. Fowler
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | | | - Klauss Mostacada
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Allison Yan
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | | | - Rae A. Stanco
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Emily D.S. Hales
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Aditi Varkey
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Wenjing Zong
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gary D. Wu
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Camila I. de Oliveira
- Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, Salvador, Brazil
| | - Patrick L. Collins
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Fernanda O. Novais
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
14
|
Suckow MA, Bolton ID, McDowell MA. Overview and Approaches for Handling of Animal Models of Leishmaniasis. Comp Med 2024; 74:148-155. [PMID: 39107941 PMCID: PMC11267445 DOI: 10.30802/aalas-cm-24-029] [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: 03/30/2024] [Revised: 05/10/2024] [Accepted: 05/19/2024] [Indexed: 08/10/2024]
Abstract
Leishmaniasis, a disease of global relevance, results from infection with the protozoan parasite, Leishmania, which is transmitted to susceptible hosts through the bite of sand flies. Multiple forms of leishmaniasis may occur, including cutaneous, mucocutaneous, and visceral. Research with animal models remains an important approach to help define basic pathophysi- ologic processes associated with infection and disease. In this regard, mice and hamsters represent the most commonly used models. The severity of leishmaniasis in animal models depends on several factors, including genotype of the host and parasite and the dose and route of administration of the parasite to the host, and severity of outcome may range from subclinical to severe illness. This review provides basic background on leishmaniasis, relevant animal models, the pathophysiology and clinical signs in animals used as models of leishmaniasis, and general approaches to mitigate risk to personnel.
Collapse
Affiliation(s)
- Mark A Suckow
- Department of Biomedical Engineering, University of Kentucky, Lexington, Kentucky
| | - Iris D Bolton
- Freimann Life Science Center, University of Notre Dame, Notre Dame, Indiana; and
| | - Mary Ann McDowell
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana
| |
Collapse
|
15
|
Tsagmo JMN, Rotureau B, Calvo Alvarez E. Animal models of neglected parasitic diseases: In vivo multimodal imaging of experimental trypanosomatid infections. Methods Cell Biol 2024; 188:205-236. [PMID: 38880525 DOI: 10.1016/bs.mcb.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
African trypanosomiases and leishmaniases are significant neglected tropical diseases (NTDs) that affect millions globally, with severe health and socio-economic consequences, especially in endemic regions. Understanding the pathogenesis and dissemination of Trypanosoma brucei and Leishmania spp. parasites within their hosts is pivotal for the development of effective interventions. Whole-body bioluminescence and fluorescence imaging systems (BLI and FLI, respectively), are powerful tools to visualize and quantify the progression and distribution of these parasites in real-time within live animal models. By combining this technology with the engineering of stable T. brucei and Leishmania spp. strains expressing luciferase and/or fluorescent proteins, crucial aspects of the infection process including the parasites' homing, the infection dynamics, the tissue tropism, or the efficacy of experimental treatments and vaccines can be deeply investigated. This methodology allows for enhanced sensitivity and resolution, elucidating previously unrecognized infection niches and dynamics. Importantly, whole-body in vivo imaging is non-invasive, enabling for longitudinal studies during the course of an infection in the same animal, thereby aligning with the "3Rs" principle of animal research. Here, we detail a protocol for the generation of dual-reporter T. brucei and L. major, and their use to infect mice and follow the spatiotemporal dynamics of infection by in vivo imaging systems. Additionally, 3D micro-computed tomography (μCT) coupled to BLI in T. brucei-infected animals is applied to gain insights into the anatomical parasite distribution. This Chapter underscores the potential of these bioimaging modalities as indispensable tools in parasitology, paving the way for novel therapeutic strategies and deeper insights into host-parasite interactions.
Collapse
Affiliation(s)
- Jean Marc Ngoune Tsagmo
- Trypanosome Transmission Group, Trypanosome Cell Biology Unit, INSERM U1201, Department of Parasites and Insect Vectors, Institut Pasteur, Université Paris Cité, Paris, France
| | - Brice Rotureau
- Trypanosome Transmission Group, Trypanosome Cell Biology Unit, INSERM U1201, Department of Parasites and Insect Vectors, Institut Pasteur, Université Paris Cité, Paris, France; Parasitology Unit, Institut Pasteur of Guinea, Conakry, Guinea
| | | |
Collapse
|
16
|
Díaz-Varela M, Sanchez-Hidalgo A, Calderon-Copete S, Tacchini V, Shipley TR, Ramírez LG, Marquis J, Fernández OL, Saravia NG, Tacchini-Cottier F. The different impact of drug-resistant Leishmania on the transcription programs activated in neutrophils. iScience 2024; 27:109773. [PMID: 38711445 PMCID: PMC11070714 DOI: 10.1016/j.isci.2024.109773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/22/2024] [Accepted: 04/15/2024] [Indexed: 05/08/2024] Open
Abstract
Drug resistance threatens the effective control of infections, including parasitic diseases such as leishmaniases. Neutrophils are essential players in antimicrobial control, but their role in drug-resistant infections is poorly understood. Here, we evaluated human neutrophil response to clinical parasite strains having distinct natural drug susceptibility. We found that Leishmania antimony drug resistance significantly altered the expression of neutrophil genes, some of them transcribed by specific neutrophil subsets. Infection with drug-resistant parasites increased the expression of detoxification pathways and reduced the production of cytokines. Among these, the chemokine CCL3 was predominantly impacted, which resulted in an impaired ability of neutrophils to attract myeloid cells. Moreover, decreased myeloid recruitment when CCL3 levels are reduced was confirmed by blocking CCL3 in a mouse model. Collectively, these findings reveal that the interplay between naturally drug-resistant parasites and neutrophils modulates the infected skin immune microenvironment, revealing a key role of neutrophils in drug resistance.
Collapse
Affiliation(s)
- Míriam Díaz-Varela
- Department of Immunobiology, WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, 1066 Epalinges, Switzerland
| | - Andrea Sanchez-Hidalgo
- Centro Internacional de Entrenamiento e Investigaciones Médicas, CIDEIM, Cali 760031, Colombia
- Universidad Icesi, Cali 760031, Colombia
| | - Sandra Calderon-Copete
- Lausanne Genomic Technologies Facility, University of Lausanne, 1015 Lausanne, Switzerland
| | - Virginie Tacchini
- Department of Immunobiology, WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, 1066 Epalinges, Switzerland
| | - Tobias R. Shipley
- Department of Immunobiology, WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, 1066 Epalinges, Switzerland
| | - Lady Giovanna Ramírez
- Centro Internacional de Entrenamiento e Investigaciones Médicas, CIDEIM, Cali 760031, Colombia
- Universidad Icesi, Cali 760031, Colombia
| | - Julien Marquis
- Lausanne Genomic Technologies Facility, University of Lausanne, 1015 Lausanne, Switzerland
| | - Olga Lucía Fernández
- Centro Internacional de Entrenamiento e Investigaciones Médicas, CIDEIM, Cali 760031, Colombia
- Universidad Icesi, Cali 760031, Colombia
| | - Nancy Gore Saravia
- Centro Internacional de Entrenamiento e Investigaciones Médicas, CIDEIM, Cali 760031, Colombia
- Universidad Icesi, Cali 760031, Colombia
| | - Fabienne Tacchini-Cottier
- Department of Immunobiology, WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, 1066 Epalinges, Switzerland
| |
Collapse
|
17
|
Ribeiro FN, de Souza TL, Menezes RC, Keidel L, dos Santos JPR, da Silva IJ, Pelajo-Machado M, Morgado FN, Porrozzi R. Anatomical Vascular Differences and Leishmania-Induced Vascular Morphological Changes Are Associated with a High Parasite Load in the Skin of Dogs Infected with Leishmania infantum. Pathogens 2024; 13:371. [PMID: 38787223 PMCID: PMC11123845 DOI: 10.3390/pathogens13050371] [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: 10/27/2023] [Revised: 12/07/2023] [Accepted: 12/14/2023] [Indexed: 05/25/2024] Open
Abstract
Canine visceral leishmaniasis (CVL), caused by the protozoan Leishmania infantum, affects several organs, including the skin. Dogs are considered the major domestic reservoir animals for leishmaniasis, and through their highly parasitized skin, they can serve as a source of infection for sandfly vectors. Therefore, studies of the skin parasite-host relationship can contribute to the understanding of the infectious dissemination processes of parasites in the dermis and help to identify targets for diagnosis and treatment. Thus, the aim of this study was to evaluate the association of anatomical vascular differences and Leishmania-induced vascular morphological changes with clinical signs and parasite load by analyzing the ear and abdominal skin from dogs naturally infected with L. infantum. Paired samples of ear and abdominal skin from L. infantum-positive dogs (n = 26) were submitted for histological and immunohistochemistry analyses. The ear skin samples showed a more intense and more diffusely distributed granulomatous inflammatory reaction, a higher number and larger diameter of blood vessels, increased parasite load, higher expression of VEGF+ (vascular endothelial growth factor) and MAC 387+ (calprotectin) recently infiltrating cells, and more intense collagen disruption compared to the abdominal skin samples. Intracellular amastigotes were observed in blood vessels and inside endothelial cells and were diffusely distributed throughout the dermis in the ear skin samples. The NOS2/MAC387+ cell ratio was lower in the ear skin samples than in those of the abdomen, suggesting that in the ear dermis, the inflammatory infiltrate was less capable of producing NO and thereby control the parasite load. Together, these findings indicate how parasites and immune cells are distributed in the skin and suggest an important role for dermal vascularization in cellular influx and thereby in parasite dissemination through the skin of naturally infected dogs.
Collapse
Affiliation(s)
- Francini N. Ribeiro
- Laboratório de Protozoologia, Instituto Oswaldo Cruz—FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (F.N.R.); (T.L.d.S.)
- Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz—FIOCRUZ, Rio de Janeiro 21040-360, Brazil
| | - Tainã L. de Souza
- Laboratório de Protozoologia, Instituto Oswaldo Cruz—FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (F.N.R.); (T.L.d.S.)
- Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz—FIOCRUZ, Rio de Janeiro 21040-360, Brazil
| | - Rodrigo C. Menezes
- Laboratório de Pesquisa Clínica em Dermatozoonoses em Animais Domésticos, Instituto Nacional de Infectologia Evandro Chagas—FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (R.C.M.); (L.K.)
| | - Lucas Keidel
- Laboratório de Pesquisa Clínica em Dermatozoonoses em Animais Domésticos, Instituto Nacional de Infectologia Evandro Chagas—FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (R.C.M.); (L.K.)
| | - João Paulo R. dos Santos
- Laboratorio de Medicina Experimental e Saúde, Instituto Oswaldo Cruz—FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (J.P.R.d.S.); (I.J.d.S.); (M.P.-M.)
| | - Igor J. da Silva
- Laboratorio de Medicina Experimental e Saúde, Instituto Oswaldo Cruz—FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (J.P.R.d.S.); (I.J.d.S.); (M.P.-M.)
| | - Marcelo Pelajo-Machado
- Laboratorio de Medicina Experimental e Saúde, Instituto Oswaldo Cruz—FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (J.P.R.d.S.); (I.J.d.S.); (M.P.-M.)
- Brazilian National Institute of Science and Technology on Neuroimmunomodulation, Instituto Oswaldo Cruz—FIOCRUZ, Rio de Janeiro 21040-360, Brazil
| | - Fernanda N. Morgado
- Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz—FIOCRUZ, Rio de Janeiro 21040-360, Brazil
| | - Renato Porrozzi
- Laboratório de Protozoologia, Instituto Oswaldo Cruz—FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (F.N.R.); (T.L.d.S.)
| |
Collapse
|
18
|
Uribe-Querol E, Rosales C. Neutrophils versus Protozoan Parasites: Plasmodium, Trichomonas, Leishmania, Trypanosoma, and Entameoba. Microorganisms 2024; 12:827. [PMID: 38674770 PMCID: PMC11051968 DOI: 10.3390/microorganisms12040827] [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: 03/10/2024] [Revised: 04/04/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Neutrophils are the most abundant polymorphonuclear granular leukocytes in human blood and are an essential part of the innate immune system. Neutrophils are efficient cells that eliminate pathogenic bacteria and fungi, but their role in dealing with protozoan parasitic infections remains controversial. At sites of protozoan parasite infections, a large number of infiltrating neutrophils is observed, suggesting that neutrophils are important cells for controlling the infection. Yet, in most cases, there is also a strong inflammatory response that can provoke tissue damage. Diseases like malaria, trichomoniasis, leishmaniasis, Chagas disease, and amoebiasis affect millions of people globally. In this review, we summarize these protozoan diseases and describe the novel view on how neutrophils are involved in protection from these parasites. Also, we present recent evidence that neutrophils play a double role in these infections participating both in control of the parasite and in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Eileen Uribe-Querol
- Laboratorio de Biología del Desarrollo, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Carlos Rosales
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
19
|
Haist KC, Gibbings SL, Jacobelli J, Mould KJ, Henson PM, Bratton DL. A LTB 4/CD11b self-amplifying loop drives pyogranuloma formation in chronic granulomatous disease. iScience 2024; 27:109589. [PMID: 38623335 PMCID: PMC11016758 DOI: 10.1016/j.isci.2024.109589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/23/2024] [Accepted: 03/25/2024] [Indexed: 04/17/2024] Open
Abstract
Sterile pyogranulomas and heightened cytokine production are hyperinflammatory hallmarks of Chronic Granulomatous Disease (CGD). Using peritoneal cells of zymosan-treated CGD (gp91phox-/-) versus wild-type (WT) mice, an ex vivo system of pyogranuloma formation was developed to determine factors involved in and consequences of recruitment of neutrophils and monocyte-derived macrophages (MoMacs). Whereas WT cells failed to aggregate, CGD cells formed aggregates containing neutrophils initially, and MoMacs recruited secondarily. LTB4 was key, as antagonizing BLT1 blocked neutrophil aggregation, but acted only indirectly on MoMac recruitment. LTB4 upregulated CD11b expression on CGD neutrophils, and the absence/blockade of CD11b inhibited LTB4 production and cell aggregation. Neutrophil-dependent MoMac recruitment was independent of MoMac Nox2 status, BLT1, CCR1, CCR2, CCR5, CXCR2, and CXCR6. As proof of concept, CD11b-deficient CGD mice developed disrupted pyogranulomas with poorly organized neutrophils and diminished recruitment of MoMacs. Importantly, the disruption of cell aggregation and pyogranuloma formation markedly reduced proinflammatory cytokine production.
Collapse
Affiliation(s)
- Kelsey C. Haist
- National Jewish Health, Department of Pediatrics, Denver, CO 80206, USA
| | | | - Jordan Jacobelli
- University of Colorado, Anschutz Medical Campus, Department of Immunology and Microbiology, Barbara Davis Research Center, Aurora, CO 80045, USA
| | - Kara J. Mould
- National Jewish Health, Department of Medicine, Denver, CO 80206, USA
- University of Colorado, Anschutz Medical Campus, Department of Pulmonary and Critical Care Medicine, Aurora, CO 80045, USA
| | - Peter M. Henson
- National Jewish Health, Department of Pediatrics, Denver, CO 80206, USA
- University of Colorado, Anschutz Medical Campus, Department of Immunology and Microbiology, Barbara Davis Research Center, Aurora, CO 80045, USA
- National Jewish Health, Department of Medicine, Denver, CO 80206, USA
- University of Colorado, Anschutz Medical Campus, Department of Pulmonary and Critical Care Medicine, Aurora, CO 80045, USA
| | - Donna L. Bratton
- National Jewish Health, Department of Pediatrics, Denver, CO 80206, USA
- University of Colorado, Anschutz Medical Campus, Department of Pediatrics, Aurora, CO 80045, USA
| |
Collapse
|
20
|
Banerjee S, Gadpayle MP, Samanta S, Dutta P, Das S, Datta R, Maiti S. Role of Macrophage PIST Protein in Regulating Leishmania major Infection. ACS Infect Dis 2024; 10:1414-1428. [PMID: 38556987 DOI: 10.1021/acsinfecdis.4c00156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
PDZ protein interacting specifically with Tc10 or PIST is a mammalian trans-Golgi resident protein that regulates subcellular sorting of plasma membrane receptors. PIST has recently emerged as a key player in regulating viral pathogenesis. Nevertheless, the involvement of PIST in parasitic infections remains unexplored. Leishmania parasites infiltrate their host macrophage cells through phagocytosis, where they subsequently multiply within the parasitophorous vacuole (PV). Host cell autophagy has been found to be important in regulating this parasite infection. Since PIST plays a pivotal role in triggering autophagy through the Beclin 1-PI3KC3 pathway, it becomes interesting to identify the status of PIST during Leishmania infection. We found that while macrophage cells are infected with Leishmania major (L. major), the expression of PIST protein remains unaltered; however, it traffics from the Golgi compartment to PV. Further, we identified that in L. major-infected macrophage cells, PIST associates with the autophagy regulatory protein Beclin 1 within the PVs; however, PIST does not interact with LC3. Reduction in PIST protein through siRNA silencing significantly increased parasite burden, whereas overexpression of PIST in macrophages restricted L. major infectivity. Together, our study reports that the macrophage PIST protein is essential in regulating L. major infectivity.
Collapse
Affiliation(s)
- Sourav Banerjee
- Department of Biological Sciences, Indian Institute of Science Education & Research Kolkata (IISER-Kolkata), Mohanpur Campus, Kolkata, West Bengal 741 246, India
| | - Mandip Pratham Gadpayle
- Department of Biological Sciences, Indian Institute of Science Education & Research Kolkata (IISER-Kolkata), Mohanpur Campus, Kolkata, West Bengal 741 246, India
| | - Suman Samanta
- Department of Biological Sciences, Indian Institute of Science Education & Research Kolkata (IISER-Kolkata), Mohanpur Campus, Kolkata, West Bengal 741 246, India
| | - Priyanka Dutta
- Department of Biological Sciences, Indian Institute of Science Education & Research Kolkata (IISER-Kolkata), Mohanpur Campus, Kolkata, West Bengal 741 246, India
| | - Swagata Das
- Department of Biological Sciences, Indian Institute of Science Education & Research Kolkata (IISER-Kolkata), Mohanpur Campus, Kolkata, West Bengal 741 246, India
| | - Rupak Datta
- Department of Biological Sciences, Indian Institute of Science Education & Research Kolkata (IISER-Kolkata), Mohanpur Campus, Kolkata, West Bengal 741 246, India
| | - Sankar Maiti
- Department of Biological Sciences, Indian Institute of Science Education & Research Kolkata (IISER-Kolkata), Mohanpur Campus, Kolkata, West Bengal 741 246, India
| |
Collapse
|
21
|
Firouzjaie F, Taghipour N, Akhavan AA, Seyyed Tabaei SJ, Rouhani S, Shirazian M, Koochaki A, Fatemi M, Mosaffa N, Moin Vaziri V. Neutrophil extracellular traps formation: effect of Leishmania major promastigotes and salivary gland homogenates of Phlebotomus papatasi in human neutrophil culture. BMC Microbiol 2024; 24:117. [PMID: 38575882 PMCID: PMC10993452 DOI: 10.1186/s12866-024-03270-z] [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: 12/25/2023] [Accepted: 03/20/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Leishmaniasis as a neglected tropical disease (NTD) is caused by the inoculation of Leishmania parasites via the bite of phlebotomine sand flies. After an infected bite, a series of innate and adaptive immune responses occurs, among which neutrophils can be mentioned as the initiators. Among the multiple functions of these fighting cells, neutrophil extracellular traps (NETs) were studied in the presence of Leishmania major promastigotes and salivary gland homogenates (SGH) of Phlebotomus papatasi alone, and in combination to mimic natural conditions of transmission. MATERIAL & METHODS The effect of L. major and SGH on NETs formation was studied in three different groups: neutrophils + SGH (NS), neutrophils + L. major (NL), neutrophils + L. major + SGH (NLS) along with negative and positive controls in 2, 4 and 6 h post-incubation. Different microscopic methods were used to visualize NETs comprising: fluorescence microscopy by Acridine Orange/ Ethidium Bromide staining, optical microscopy by Giemsa staining and scanning electron microscopy. In addition, the expression level of three different genes NE, MPO and MMP9 was evaluated by Real-Time PCR. RESULTS All three microscopical methods revealed similar results, as in NS group, chromatin extrusion as a sign of NETosis, was not very evident in each three time points; but, in NL and especially NLS group, more NETosis was observed and the interaction between neutrophils and promastigotes in NL and also with saliva in NLS group, gradually increased over times. Real-time reveals that, the expression of MPO, NE and MMP9 genes increased during 2 and 4 h after exposure, and then decreased at 6 h in most groups. CONCLUSION Hence, it was determined that the simultaneous presence of parasite and saliva in NLS group has a greater impact on the formation of NETs compared to NL and NS groups.
Collapse
Affiliation(s)
- Fahimeh Firouzjaie
- Department of Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Niloofar Taghipour
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Ahmad Akhavan
- Department of Vector Biology and Control of Diseases, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyyed Javad Seyyed Tabaei
- Department of Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soheila Rouhani
- Department of Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Shirazian
- Department of Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ameneh Koochaki
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahboubeh Fatemi
- Department of Vector Biology and Control of Diseases, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Nariman Mosaffa
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Vahideh Moin Vaziri
- Department of Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Seyed N, Taheri T, Rafati S. Live attenuated-nonpathogenic Leishmania and DNA structures as promising vaccine platforms against leishmaniasis: innovations can make waves. Front Microbiol 2024; 15:1326369. [PMID: 38633699 PMCID: PMC11021776 DOI: 10.3389/fmicb.2024.1326369] [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: 10/28/2023] [Accepted: 03/12/2024] [Indexed: 04/19/2024] Open
Abstract
Leishmaniasis is a vector-borne disease caused by the protozoan parasite of Leishmania genus and is a complex disease affecting mostly tropical regions of the world. Unfortunately, despite the extensive effort made, there is no vaccine available for human use. Undoubtedly, a comprehensive understanding of the host-vector-parasite interaction is substantial for developing an effective prophylactic vaccine. Recently the role of sandfly saliva on disease progression has been uncovered which can make a substantial contribution in vaccine design. In this review we try to focus on the strategies that most probably meet the prerequisites of vaccine development (based on the current understandings) including live attenuated/non-pathogenic and subunit DNA vaccines. Innovative approaches such as reverse genetics, CRISP/R-Cas9 and antibiotic-free selection are now available to promisingly compensate for intrinsic drawbacks associated with these platforms. Our main goal is to call more attention toward the prerequisites of effective vaccine development while controlling the disease outspread is a substantial need.
Collapse
Affiliation(s)
- Negar Seyed
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | | | | |
Collapse
|
23
|
Morales-Primo AU, Becker I, Pedraza-Zamora CP, Zamora-Chimal J. Th17 Cell and Inflammatory Infiltrate Interactions in Cutaneous Leishmaniasis: Unraveling Immunopathogenic Mechanisms. Immune Netw 2024; 24:e14. [PMID: 38725676 PMCID: PMC11076297 DOI: 10.4110/in.2024.24.e14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/26/2024] [Accepted: 03/06/2024] [Indexed: 05/12/2024] Open
Abstract
The inflammatory response during cutaneous leishmaniasis (CL) involves immune and non-immune cell cooperation to contain and eliminate Leishmania parasites. The orchestration of these responses is coordinated primarily by CD4+ T cells; however, the disease outcome depends on the Th cell predominant phenotype. Although Th1 and Th2 phenotypes are the most addressed as steers for the resolution or perpetuation of the disease, Th17 cell activities, especially IL-17 release, are recognized to be vital during CL development. Th17 cells perform vital functions during both acute and chronic phases of CL. Overall, Th17 cells induce the migration of phagocytes (neutrophils, macrophages) to the infection site and CD8+ T cells and NK cell activation. They also provoke granzyme and perforin secretion from CD8+ T cells, macrophage differentiation towards an M2 phenotype, and expansion of B and Treg cells. Likewise, immune cells from the inflammatory infiltrate have modulatory activities over Th17 cells involving their differentiation from naive CD4+ T cells and further expansion by generating a microenvironment rich in optimal cytokines such as IL-1β, TGF-β, IL-6, and IL-21. Th17 cell activities and synergies are crucial for the resistance of the infection during the early and acute stages; however, if unchecked, Th17 cells might lead to a chronic stage. This review discusses the synergies between Th17 cells and the inflammatory infiltrate and how these interactions might destine the course of CL.
Collapse
Affiliation(s)
- Abraham U. Morales-Primo
- Laboratorio de Inmunoparasitología, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Mexico City 06720, México
| | - Ingeborg Becker
- Laboratorio de Inmunoparasitología, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Mexico City 06720, México
| | - Claudia Patricia Pedraza-Zamora
- Laboratorio de Biología Periodontal y Tejidos Mineralizados, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Mexico City 04510, México
| | - Jaime Zamora-Chimal
- Laboratorio de Inmunoparasitología, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Mexico City 06720, México
| |
Collapse
|
24
|
Roy K, Ghosh S, Karan M, Karmakar S, Nath S, Das B, Paul S, Mandal P, Ray M, Das M, Mukherjee S, Dey S, Pal C. Activation of neutrophils excels the therapeutic potential of Mycobacterium indicus pranii and heat-induced promastigotes against antimony-resistant Leishmania donovani infection. Scand J Immunol 2024; 99:e13350. [PMID: 39008005 DOI: 10.1111/sji.13350] [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: 02/27/2023] [Revised: 11/08/2023] [Accepted: 12/22/2023] [Indexed: 07/16/2024]
Abstract
Repurposing drugs and adjuvants is an attractive choice of present therapy that reduces the substantial costs, chances of failure, and systemic toxicity. Mycobacterium indicus pranii was originally developed as a leprosy vaccine but later has been found effective against Leishmania donovani infection. To extend our earlier study, here we reported the immunotherapeutic modulation of the splenic and circulatory neutrophils in favour of hosts as neutrophils actually serve as the pro-parasitic portable shelter to extend the Leishmania infection specifically during the early entry into the hosts' circulation. We targeted to disrupt this early pro-parasitic incidence by the therapeutic combination of M. indicus pranii and heat-induced promastigotes against antimony-resistant L. donovani infection. The combination therapy induced the functional expansion of CD11b+Ly6CintLy6Ghi neutrophils both in the post-infected spleen, and also in the circulation of post-treated animals followed by the immediate Leishmania infection. More importantly, the enhanced expression of MHC-II, phagocytic uptake of the parasites by the circulatory neutrophils as well as the oxidative burst were induced that limited the chances of the very early establishment of the infection. The enhanced expression of pro-inflammatory cytokines, like IL-1α and TNF-α indicated resistance to the parasite-mediated takeover of the neutrophils, as these cytokines are critical for the activation of T cell-mediated immunity and host-protective responses. Additionally, the induction of essential transcription factors and cytokines for early granulocytic lineage commitment suggests that the strategy not only contributed to the peripheral activation of the neutrophils but also promoted granulopoiesis in the bone marrow.
Collapse
Affiliation(s)
- Kamalika Roy
- Cellular Immunology and Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, West Bengal, India
| | - Sanhita Ghosh
- Cellular Immunology and Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, West Bengal, India
| | - Mintu Karan
- Cellular Immunology and Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, West Bengal, India
| | - Suman Karmakar
- Cellular Immunology and Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, West Bengal, India
| | - Supriya Nath
- Cellular Immunology and Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, West Bengal, India
| | - Bedanta Das
- Cellular Immunology and Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, West Bengal, India
| | - Sharmistha Paul
- Cellular Immunology and Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, West Bengal, India
| | - Pritam Mandal
- Cellular Immunology and Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, West Bengal, India
| | - Monalisa Ray
- Cellular Immunology and Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, West Bengal, India
| | - Mousumi Das
- Cellular Immunology and Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, West Bengal, India
| | - Soumyadip Mukherjee
- Cellular Immunology and Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, West Bengal, India
| | - Somaditya Dey
- Cellular Immunology and Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, West Bengal, India
| | - Chiranjib Pal
- Cellular Immunology and Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, West Bengal, India
| |
Collapse
|
25
|
Peyvandi S, Lan Q, Chabloz A, Prével F, La Torre YH, Ives A, Tacchini-Cottier F. The β-Carboline Harmine Has a Protective Immunomodulatory Role in Nonhealing Cutaneous Leishmaniasis. J Invest Dermatol 2024; 144:862-873.e4. [PMID: 37852357 DOI: 10.1016/j.jid.2023.09.280] [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: 07/11/2023] [Revised: 09/15/2023] [Accepted: 09/28/2023] [Indexed: 10/20/2023]
Abstract
Cutaneous leishmaniasis affects 1 million people worldwide annually. Although conventional treatments primarily target the parasite, there is growing interest in host immune modulation. In this study, we investigated the impact of synthetic β-carboline harmine (ACB1801), previously shown to be immunoregulatory in cancer, on the pathology caused by a drug-resistant Leishmania major strain causing persistent cutaneous lesions. Exposure to ACB1801 in vitro had a modest impact on parasite burden within host macrophages. Moreover, it significantly increased major histocompatibility complex II and costimulatory molecule expression on infected dendritic cells, suggesting an enhanced immune response. In vivo, ACB1801 monotherapy led to a substantial reduction in lesion development and parasite burden in infected C57BL/6 mice, comparable with efficacy of amphotericin B. Transcriptomics analysis further supported ACB1801 immunomodulatory effects, revealing an enrichment of TNF-α, IFN-γ, and major histocompatibility complex II antigen presentation signatures in the draining lymph nodes of treated mice. Flow cytometry analysis confirmed an increased frequency (1.5×) of protective CD4+IFN-γ+TNF-α+ T cells and a decreased frequency (2×) in suppressive IL-10+FoxP3- T cells at the site of infection and in draining lymph nodes. In addition, ACB1801 downregulated the aryl hydrocarbon receptor signaling, known to enhance immunosuppressive cytokines. Thus, these results suggest a potential use for ACB1801 alone or in combination therapy for cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Sanam Peyvandi
- Department of Immunobiology, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland
| | - Qiang Lan
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | | | - Florence Prével
- Department of Immunobiology, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland
| | - Yazmin Hauyon La Torre
- Department of Immunobiology, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland
| | | | - Fabienne Tacchini-Cottier
- Department of Immunobiology, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland.
| |
Collapse
|
26
|
Rizo-Téllez SA, Filep JG. Beyond host defense and tissue injury: the emerging role of neutrophils in tissue repair. Am J Physiol Cell Physiol 2024; 326:C661-C683. [PMID: 38189129 PMCID: PMC11193466 DOI: 10.1152/ajpcell.00652.2023] [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: 11/29/2023] [Revised: 12/31/2023] [Accepted: 12/31/2023] [Indexed: 01/09/2024]
Abstract
Neutrophils, the most abundant immune cells in human blood, play a fundamental role in host defense against invading pathogens and tissue injury. Neutrophils carry potentially lethal weaponry to the affected site. Inadvertent and perpetual neutrophil activation could lead to nonresolving inflammation and tissue damage, a unifying mechanism of many common diseases. The prevailing view emphasizes the dichotomy of their function, host defense versus tissue damage. However, tissue injury may also persist during neutropenia, which is associated with disease severity and poor outcome. Numerous studies highlight neutrophil phenotypic heterogeneity and functional versatility, indicating that neutrophils play more complex roles than previously thought. Emerging evidence indicates that neutrophils actively orchestrate resolution of inflammation and tissue repair and facilitate return to homeostasis. Thus, neutrophils mobilize multiple mechanisms to limit the inflammatory reaction, assure debris removal, matrix remodeling, cytokine scavenging, macrophage reprogramming, and angiogenesis. In this review, we will summarize the homeostatic and tissue-reparative functions and mechanisms of neutrophils across organs. We will also discuss how the healing power of neutrophils might be harnessed to develop novel resolution and repair-promoting therapies while maintaining their defense functions.
Collapse
Affiliation(s)
- Salma A Rizo-Téllez
- Department of Pathology and Cell Biology, University of Montreal and Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
| | - János G Filep
- Department of Pathology and Cell Biology, University of Montreal and Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
| |
Collapse
|
27
|
Volkmar K, Jaedtka M, Baars I, Walber B, Philipp MS, Bagola K, Müller AJ, Heine H, van Zandbergen G. Investigating pyroptosis as a mechanism of L. major cell-to-cell spread in the human BLaER1 infection model. Mol Microbiol 2024; 121:453-469. [PMID: 37612878 DOI: 10.1111/mmi.15142] [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: 03/14/2023] [Revised: 07/04/2023] [Accepted: 08/03/2023] [Indexed: 08/25/2023]
Abstract
Leishmania is the causative agent of the tropical neglected disease leishmaniasis and infects macrophages as its definitive host cell. In order to sustain and propagate infections, Leishmania parasites have to complete cycles of exit and re-infection. Yet, the mechanism driving the parasite spread to other cells remains unclear. Recent studies reported pro-inflammatory monocytes as replicative niche of Leishmania major and showed prolonged expression of IL-1β at the site of infection, indicating an activation of the NLRP3 inflammasome and pointing toward pyroptosis as a possible mechanism of parasite spread. To address the species-specific inflammasome activation of human cells, we characterized the BLaER1 monocytes as a model for L. major infection. We found that BLaER1 monocytes support infection and activation by Leishmania parasites to the same extent as primary human macrophages. Harnessing the possibilities of this infection model, we first showed that BLaER1 GSDMD-/- cells, which carry a deletion of the pore-forming protein gasdermin D, are more resistant to pyroptotic cell death and, concomitantly, display a strongly delayed release of intracellular parasite. Using that knockout in a co-incubation assay in comparison with wild-type BLaER1 cells, we demonstrate that impairment of the pyroptosis pathway leads to lower rates of parasite spread to new host cells, thus, implicating pyroptotic cell death as a possible exit mechanism of L. major in pro-inflammatory microenvironments.
Collapse
Affiliation(s)
- Kerren Volkmar
- Division of Immunology, Paul-Ehrlich-Institut Langen, Langen, Germany
- Institute for Immunology, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Moritz Jaedtka
- Division of Immunology, Paul-Ehrlich-Institut Langen, Langen, Germany
- Institute for Immunology, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Iris Baars
- Experimental Immunodynamics, Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Bianca Walber
- Division of Immunology, Paul-Ehrlich-Institut Langen, Langen, Germany
| | | | - Katrin Bagola
- Division of Immunology, Paul-Ehrlich-Institut Langen, Langen, Germany
| | - Andreas J Müller
- Experimental Immunodynamics, Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Holger Heine
- Division of Innate Immunity, Research Center Borstel-Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Ger van Zandbergen
- Division of Immunology, Paul-Ehrlich-Institut Langen, Langen, Germany
- Institute for Immunology, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
28
|
Uderhardt S, Neag G, Germain RN. Dynamic Multiplex Tissue Imaging in Inflammation Research. ANNUAL REVIEW OF PATHOLOGY 2024; 19:43-67. [PMID: 37722698 DOI: 10.1146/annurev-pathmechdis-070323-124158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/20/2023]
Abstract
Inflammation is a highly dynamic process with immune cells that continuously interact with each other and parenchymal components as they migrate through tissue. The dynamic cellular responses and interaction patterns are a function of the complex tissue environment that cannot be fully reconstructed ex vivo, making it necessary to assess cell dynamics and changing spatial patterning in vivo. These dynamics often play out deep within tissues, requiring the optical focus to be placed far below the surface of an opaque organ. With the emergence of commercially available two-photon excitation lasers that can be combined with existing imaging systems, new avenues for imaging deep tissues over long periods of time have become available. We discuss a selected subset of studies illustrating how two-photon microscopy (2PM) has helped to relate the dynamics of immune cells to their in situ function and to understand the molecular patterns that govern their behavior in vivo. We also review some key practical aspects of 2PM methods and point out issues that can confound the results, so that readers can better evaluate the reliability of conclusions drawn using this technology.
Collapse
Affiliation(s)
- Stefan Uderhardt
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
- Exploratory Research Unit, Optical Imaging Competence Centre, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Georgiana Neag
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
- Exploratory Research Unit, Optical Imaging Competence Centre, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Ronald N Germain
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Center for Advanced Tissue Imaging (CAT-I), National Institute of Allergy and Infectious Diseases and National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA;
| |
Collapse
|
29
|
Muhammed Hassan G, Zuhair Ali H, Muhammed Hussein W. Evaluation of IL-8, nitric oxide and macrophage inhibitory factor as clinical circulatory markers in patients with cutaneous leishmaniasis, before and during sodium stibogluconate treatment. Cytokine 2024; 173:156450. [PMID: 37988922 DOI: 10.1016/j.cyto.2023.156450] [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: 10/06/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 11/23/2023]
Abstract
The clinical spectrum of cutaneous leishmaniasis (CL), an intracellular parasitic pathogen, ranges from a single sore healing to chronic crusty lesions with a manifestation of treatment resistance. The complicated interaction between Leishmania bodies and the early immune response, including innate and adaptive mechanisms, determines the evolution of nodules. This study examined the levels of the chemoattractant interleukin 8 (IL-8), pro-inflammatory nitric oxide (NO), and immunoregulatory macrophage inhibitory factor (MIF) in the serum of subjects recently diagnosed with cutaneous leishmaniasis, in parallel with patients being monitored during consecutive sodium stibogluconate (Pentostam) treatment. A total of 161 serum samples of newly diagnosed individuals and patients undergoing pentostam injections were collected form an endemic area of Diyala, east central of Iraq. Sandwich ELISA was used to measure the level of IL-8, NO and MIF in the studied groups. Results of circulatory markers levels showed a considerable difference in all groups, with IL-8 being exceptionally higher in the first two groups of pretreated and dose-1 (191.5, 273.64) pg/ml respectively, while NO was found to be lower than in control subjects, particularly in the pretreated group (12.08 µmol/L) and MIF level was significantly higher in the pretreated group, which was (7.18 pg/ml). These findings can provide insights for distinction of disease phase and monitoring treatment efficacy along consecutive dosages, particularly in populations where CL is endemic.
Collapse
Affiliation(s)
- Ghuffran Muhammed Hassan
- Deptartment of Biology, College of Science, University of Baghdad, Al-Jaderiya Campus, Baghdad 10071, Iraq
| | - Hayder Zuhair Ali
- Deptartment of Biology, College of Science, University of Baghdad, Al-Jaderiya Campus, Baghdad 10071, Iraq.
| | | |
Collapse
|
30
|
Zayats R, Mou Z, Yazdanpanah A, Gupta G, Lopez P, Nayar D, Koh WH, Uzonna JE, Murooka TT. Antigen recognition reinforces regulatory T cell mediated Leishmania major persistence. Nat Commun 2023; 14:8449. [PMID: 38114497 PMCID: PMC10730873 DOI: 10.1038/s41467-023-44297-6] [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: 06/13/2023] [Accepted: 12/07/2023] [Indexed: 12/21/2023] Open
Abstract
Cutaneous Leishmania major infection elicits a rapid T cell response that is insufficient to clear residually infected cells, possibly due to the accumulation of regulatory T cells in healed skin. Here, we used Leishmania-specific TCR transgenic mice as a sensitive tool to characterize parasite-specific effector and immunosuppressive responses in vivo using two-photon microscopy. We show that Leishmania-specific Tregs displayed higher suppressive activity compared to polyclonal Tregs, that was mediated through IL-10 and not through disrupting cell-cell contacts or antigen presentation. In vivo expansion of endogenous Leishmania-specific Tregs resulted in disease reactivation that was also IL-10 dependent. Interestingly, lack of Treg expansion that recognized the immunodominant Leishmania peptide PEPCK was sufficient to restore robust effector Th1 responses and resulted in parasite control exclusively in male hosts. Our data suggest a stochastic model of Leishmania major persistence in skin, where cellular factors that control parasite numbers are counterbalanced by Leishmania-specific Tregs that facilitate parasite persistence.
Collapse
Affiliation(s)
- Romaniya Zayats
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Zhirong Mou
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Atta Yazdanpanah
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Gaurav Gupta
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Paul Lopez
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Deesha Nayar
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Wan H Koh
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Jude E Uzonna
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
- Department of Medical Microbiology and Infectious Diseases, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
| | - Thomas T Murooka
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
- Department of Medical Microbiology and Infectious Diseases, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
31
|
Silva Pereira S, Brás D, Porqueddu T, Nascimento AM, De Niz M. Investigation of Trypanosoma-induced vascular damage sheds insights into Trypanosoma vivax sequestration. Cell Surf 2023; 10:100113. [PMID: 37954640 PMCID: PMC10632540 DOI: 10.1016/j.tcsw.2023.100113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/22/2023] [Accepted: 10/22/2023] [Indexed: 11/14/2023] Open
Abstract
Multiple blood-borne pathogens infecting mammals establish close interactions with the host vascular endothelium as part of their life cycles. In this work, we investigate differences in the interactions of three Trypanosoma species: T. brucei, T. congolense and T. vivax with the blood vasculature. Infection with these species results in vastly different pathologies, including different effects on vascular homeostasis, such as changes in vascular permeability and microhemorrhages. While all three species are extracellular parasites, T. congolense is strictly intravascular, while T. brucei is capable of surviving both extra- and intravascularly. Our knowledge regarding T. vivax tropism and its capacity of migration across the vascular endothelium is unknown. In this work, we show for the first time that T. vivax parasites sequester to the vascular endothelium of most organs, and that, like T. congolense, T. vivax Y486 is largely incapable of extravasation. Infection with this parasite species results in a unique effect on vascular endothelium receptors including general downregulation of ICAM1 and ESAM, and upregulation of VCAM1, CD36 and E-selectin. Our findings on the differences between the two sequestering species (T. congolense and T. vivax) and the non-sequestering, but extravasating, T. brucei raise important questions on the relevance of sequestration to the parasite's survival in the mammalian host, and the evolutionary relevance of both sequestration and extravasation.
Collapse
Affiliation(s)
- Sara Silva Pereira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Católica Biomedical Research Centre, Católica Medical School, Universidade Católica Portuguesa, Lisbon, Portugal
| | - Daniela Brás
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Teresa Porqueddu
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Ana M. Nascimento
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Mariana De Niz
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Center for Advanced Microscopy and Nikon Imaging Center, Northwestern University, Chicago, USA
| |
Collapse
|
32
|
Cibir Z, Hassel J, Sonneck J, Kowitz L, Beer A, Kraus A, Hallekamp G, Rosenkranz M, Raffelberg P, Olfen S, Smilowski K, Burkard R, Helfrich I, Tuz AA, Singh V, Ghosh S, Sickmann A, Klebl AK, Eickhoff JE, Klebl B, Seidl K, Chen J, Grabmaier A, Viga R, Gunzer M. ComplexEye: a multi-lens array microscope for high-throughput embedded immune cell migration analysis. Nat Commun 2023; 14:8103. [PMID: 38081825 PMCID: PMC10713721 DOI: 10.1038/s41467-023-43765-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Autonomous migration is essential for the function of immune cells such as neutrophils and plays an important role in numerous diseases. The ability to routinely measure or target it would offer a wealth of clinical applications. Video microscopy of live cells is ideal for migration analysis, but cannot be performed at sufficiently high-throughput (HT). Here we introduce ComplexEye, an array microscope with 16 independent aberration-corrected glass lenses spaced at the pitch of a 96-well plate to produce high-resolution movies of migrating cells. With the system, we enable HT migration analysis of immune cells in 96- and 384-well plates with very energy-efficient performance. We demonstrate that the system can measure multiple clinical samples simultaneously. Furthermore, we screen 1000 compounds and identify 17 modifiers of migration in human neutrophils in just 4 days, a task that requires 60-times longer with a conventional video microscope. ComplexEye thus opens the field of phenotypic HT migration screens and enables routine migration analysis for the clinical setting.
Collapse
Affiliation(s)
- Zülal Cibir
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Jacqueline Hassel
- Department of Electronic Components and Circuits, University of Duisburg-Essen, Duisburg, Germany
| | - Justin Sonneck
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
- Faculty of Computer Science, Ruhr-Universität Bochum, 44801, Bochum, Germany
| | - Lennart Kowitz
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Alexander Beer
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Andreas Kraus
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Gabriel Hallekamp
- Department of Electronic Components and Circuits, University of Duisburg-Essen, Duisburg, Germany
| | - Martin Rosenkranz
- Department of Electronic Components and Circuits, University of Duisburg-Essen, Duisburg, Germany
| | - Pascal Raffelberg
- Department of Electronic Components and Circuits, University of Duisburg-Essen, Duisburg, Germany
| | - Sven Olfen
- Department of Electronic Components and Circuits, University of Duisburg-Essen, Duisburg, Germany
| | - Kamil Smilowski
- Department of Electronic Components and Circuits, University of Duisburg-Essen, Duisburg, Germany
| | - Roman Burkard
- Department of Electronic Components and Circuits, University of Duisburg-Essen, Duisburg, Germany
| | - Iris Helfrich
- Department of Dermatology and Allergology, Medical Faculty of the Ludwig Maximilian University of Munich, Munich, Germany
| | - Ali Ata Tuz
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Vikramjeet Singh
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Susmita Ghosh
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801, Bochum, Germany
- Department of Chemistry, College of Physical Sciences, University of Aberdeen, AB24 3FX, Aberdeen, UK
| | | | | | - Bert Klebl
- Lead Discovery Center GmbH, Dortmund, Germany
| | - Karsten Seidl
- Department of Electronic Components and Circuits, University of Duisburg-Essen, Duisburg, Germany
| | - Jianxu Chen
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Anton Grabmaier
- Department of Electronic Components and Circuits, University of Duisburg-Essen, Duisburg, Germany
| | - Reinhard Viga
- Department of Electronic Components and Circuits, University of Duisburg-Essen, Duisburg, Germany.
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany.
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany.
| |
Collapse
|
33
|
Fowler EA, Amorim CF, Mostacada K, Yan A, Sacramento LA, Stanco RA, Hales EDS, Varkey A, Zong W, Wu GD, de Oliveira CI, Collins PL, Novais FO. Pathogenic CD8 T cell responses are driven by neutrophil-mediated hypoxia in cutaneous leishmaniasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.18.562926. [PMID: 37904953 PMCID: PMC10614852 DOI: 10.1101/2023.10.18.562926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Cutaneous leishmaniasis caused by Leishmania parasites exhibits a wide range of clinical manifestations. Although parasites influence disease severity, cytolytic CD8 T cell responses mediate disease. While these responses originate in the lymph node, we find that expression of the cytolytic effector molecule granzyme B is restricted to lesional CD8 T cells in Leishmania - infected mice, suggesting that local cues within inflamed skin induce cytolytic function. Expression of Blimp-1 ( Prdm1 ), a transcription factor necessary for cytolytic CD8 T cell differentiation, is driven by hypoxia within the inflamed skin. Hypoxia is further enhanced by the recruitment of neutrophils that consume oxygen to produce reactive oxygen species, ultimately increasing granzyme B expression in CD8 T cells. Importantly, lesions from cutaneous leishmaniasis patients exhibit hypoxia transcription signatures that correlate with the presence of neutrophils. Thus, targeting hypoxia-driven signals that support local differentiation of cytolytic CD8 T cells may improve the prognosis for patients with cutaneous leishmaniasis, as well as other inflammatory skin diseases where cytolytic CD8 T cells contribute to pathogenesis.
Collapse
|
34
|
Mohammad-Rafiei F, Moadab F, Mahmoudi A, Navashenaq JG, Gheibihayat SM. Efferocytosis: a double-edged sword in microbial immunity. Arch Microbiol 2023; 205:370. [PMID: 37925389 DOI: 10.1007/s00203-023-03704-8] [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: 09/03/2023] [Revised: 09/30/2023] [Accepted: 10/10/2023] [Indexed: 11/06/2023]
Abstract
Efferocytosis is characterized as the rapid and efficient process by which dying or dead cells are removed. This type of clearance is initiated via "find-me" signals, and then, carries on by "eat-me" and "don't-eat-me" ones. Efferocytosis has a critical role to play in tissue homeostasis and innate immunity. However, some evidence suggests it as a double-edged sword in microbial immunity. In other words, some pathogens have degraded efferocytosis by employing efferocytic mechanisms to bypass innate immune detection and promote infection, despite the function of this process for the control and clearance of pathogens. In this review, the efferocytosis mechanisms from the recognition of dying cells to phagocytic engulfment are initially presented, and then, its diverse roles in inflammation and immunity are highlighted. In this case, much focus is also laid on some bacterial, viral, and parasitic infections caused by Mycobacterium tuberculosis (M. tb), Mycobacterium marinum (M. marinum), Listeria monocytogenes (L. monocytogenes), Chlamydia pneumoniae (CP), Klebsiella pneumoniae (KP), Influenza A virus (IAV), human immunodeficiency virus (HIV), and Leishmania, respectively.
Collapse
Affiliation(s)
- Fatemeh Mohammad-Rafiei
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Moadab
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, USA
| | - Ali Mahmoudi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | | | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
35
|
Bousso P, Grandjean CL. Immunomodulation under the lens of real-time in vivo imaging. Eur J Immunol 2023; 53:e2249921. [PMID: 37051691 DOI: 10.1002/eji.202249921] [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: 07/27/2022] [Revised: 11/16/2022] [Accepted: 04/11/2023] [Indexed: 04/14/2023]
Abstract
Modulation of cells and molecules of the immune system not only represents a major opportunity to treat a variety of diseases including infections, cancer, autoimmune, and inflammatory disorders but could also help understand the intricacies of immune responses. A detailed mechanistic understanding of how a specific immune intervention may provide clinical benefit is essential for the rational design of efficient immunomodulators. Visualizing the impact of immunomodulation in real-time and in vivo has emerged as an important approach to achieve this goal. In this review, we aim to illustrate how multiphoton intravital imaging has helped clarify the mode of action of immunomodulatory strategies such as antibodies or cell therapies. We also discuss how optogenetics combined with imaging will further help manipulate and precisely understand immunomodulatory pathways. Combined with other single-cell technologies, in vivo dynamic imaging has therefore a major potential for guiding preclinical development of immunomodulatory drugs.
Collapse
Affiliation(s)
- Philippe Bousso
- Dynamics of Immune Responses Unit, Institut Pasteur, INSERM U1223, Université de Paris Cité, Paris, France
| | - Capucine L Grandjean
- Dynamics of Immune Responses Unit, Institut Pasteur, INSERM U1223, Université de Paris Cité, Paris, France
| |
Collapse
|
36
|
Ihedioha OC, Sivakoses A, Beverley SM, McMahon-Pratt D, Bothwell ALM. Leishmania major-derived lipophosphoglycan influences the host's early immune response by inducing platelet activation and DKK1 production via TLR1/2. Front Immunol 2023; 14:1257046. [PMID: 37885890 PMCID: PMC10598878 DOI: 10.3389/fimmu.2023.1257046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/25/2023] [Indexed: 10/28/2023] Open
Abstract
Background Platelets are rapidly deployed to infection sites and respond to pathogenic molecules via pattern recognition receptors (TLR, NLRP). Dickkopf1 (DKK1) is a quintessential Wnt antagonist produced by a variety of cell types including platelets, endothelial cells, and is known to modulate pro-inflammatory responses in infectious diseases and cancer. Moreover, DKK1 is critical for forming leukocyte-platelet aggregates and induction of type 2 cell-mediated immune responses. Our previous publication showed activated platelets release DKK1 following Leishmania major recognition. Results Here we probed the role of the key surface virulence glycoconjugate lipophosphoglycan (LPG), on DKK1 production using null mutants deficient in LPG synthesis (Δlpg1- and Δlpg2-). Leishmania-induced DKK1 production was reduced to control levels in the absence of LPG in both mutants and was restored upon re-expression of the cognate LPG1 or LPG2 genes. Furthermore, the formation of leukocyte-platelet aggregates was dependent on LPG. LPG mediated platelet activation and DKK1 production occurs through TLR1/2. Conclusion Thus, LPG is a key virulence factor that induces DKK1 production from activated platelets, and the circulating DKK1 promotes Th2 cell polarization. This suggests that LPG-activated platelets can drive innate and adaptive immune responses to Leishmania infection.
Collapse
Affiliation(s)
- Olivia C. Ihedioha
- 1Department of Immunobiology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Anutr Sivakoses
- 1Department of Immunobiology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Stephen M. Beverley
- Department of Molecular Microbiology, Washington University School of Medicine in St Louis, St. Louis, MI, United States
| | - Diane McMahon-Pratt
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, United States
| | - Alfred L. M. Bothwell
- 1Department of Immunobiology, College of Medicine, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
37
|
Petersen J, Du W, Adkisson C, Gravekamp C, Oktay MH, Condeelis J, Panarelli NC, McAuliffe JC, Entenberg D. Stabilized Window for Intravital Imaging of the Murine Pancreas. J Vis Exp 2023:10.3791/65498. [PMID: 37870314 PMCID: PMC10731889 DOI: 10.3791/65498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023] Open
Abstract
The physiology and pathophysiology of the pancreas are complex. Diseases of the pancreas, such as pancreatitis and pancreatic adenocarcinoma (PDAC) have high morbidity and mortality. Intravital imaging (IVI) is a powerful technique enabling the high-resolution imaging of tissues in both healthy and diseased states, allowing for real-time observation of cell dynamics. IVI of the murine pancreas presents significant challenges due to the deep visceral and compliant nature of the organ, which make it highly prone to damage and motion artifacts. Described here is the process of implantation of the Stabilized Window for Intravital imaging of the murine Pancreas (SWIP). The SWIP allows IVI of the murine pancreas in normal healthy states, during the transformation from the healthy pancreas to acute pancreatitis induced by cerulein, and in malignant states such as pancreatic tumors. In conjunction with genetically labeled cells or the administration of fluorescent dyes, the SWIP enables the measurement of single-cell and subcellular dynamics (including single-cell and collective migration) as well as serial imaging of the same region of interest over multiple days. The ability to capture tumor cell migration is of particular importance as the primary cause of cancer-related mortality in PDAC is the overwhelming metastatic burden. Understanding the physiological dynamics of metastasis in PDAC is a critical unmet need and crucial for improving patient prognosis. Overall, the SWIP provides improved imaging stability and expands the application of IVI in the healthy pancreas and malignant pancreas diseases.
Collapse
Affiliation(s)
- Jakeb Petersen
- Department of Surgery, Einstein College of Medicine / Montefiore Medical Center; Department of Pathology, Einstein College of Medicine / Montefiore Medical Center; Integrated Imaging Program for Cancer Research, Einstein College of Medicine / Montefiore Medical Center
| | - Wei Du
- Department of Anatomy and Structural Biology, Einstein College of Medicine / Montefiore Medical Center; Breast Center, Peking University People's Hospital
| | - Christian Adkisson
- Department of Surgery, Einstein College of Medicine / Montefiore Medical Center; Department of Anatomy and Structural Biology, Einstein College of Medicine / Montefiore Medical Center
| | - Claudia Gravekamp
- Department of Microbiology & Immunology, Einstein College of Medicine / Montefiore Medical Center; Montefiore Einstein Cancer Center, Einstein College of Medicine / Montefiore Medical Center
| | - Maja H Oktay
- Department of Pathology, Einstein College of Medicine / Montefiore Medical Center; Integrated Imaging Program for Cancer Research, Einstein College of Medicine / Montefiore Medical Center; Department of Anatomy and Structural Biology, Einstein College of Medicine / Montefiore Medical Center; Montefiore Einstein Cancer Center, Einstein College of Medicine / Montefiore Medical Center; Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine / Montefiore Medical Center; Gruss-Lipper Biophotonics Center, Einstein College of Medicine / Montefiore Medical Center
| | - John Condeelis
- Department of Surgery, Einstein College of Medicine / Montefiore Medical Center; Integrated Imaging Program for Cancer Research, Einstein College of Medicine / Montefiore Medical Center; Department of Anatomy and Structural Biology, Einstein College of Medicine / Montefiore Medical Center; Montefiore Einstein Cancer Center, Einstein College of Medicine / Montefiore Medical Center; Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine / Montefiore Medical Center; Gruss-Lipper Biophotonics Center, Einstein College of Medicine / Montefiore Medical Center; Department of Cell Biology, Einstein College of Medicine / Montefiore Medical Center
| | - Nicole C Panarelli
- Department of Pathology, Einstein College of Medicine / Montefiore Medical Center; Integrated Imaging Program for Cancer Research, Einstein College of Medicine / Montefiore Medical Center; Montefiore Einstein Cancer Center, Einstein College of Medicine / Montefiore Medical Center
| | - John C McAuliffe
- Department of Surgery, Einstein College of Medicine / Montefiore Medical Center; Department of Pathology, Einstein College of Medicine / Montefiore Medical Center; Integrated Imaging Program for Cancer Research, Einstein College of Medicine / Montefiore Medical Center; Montefiore Einstein Cancer Center, Einstein College of Medicine / Montefiore Medical Center; Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine / Montefiore Medical Center
| | - David Entenberg
- Department of Pathology, Einstein College of Medicine / Montefiore Medical Center; Integrated Imaging Program for Cancer Research, Einstein College of Medicine / Montefiore Medical Center; Department of Anatomy and Structural Biology, Einstein College of Medicine / Montefiore Medical Center; Montefiore Einstein Cancer Center, Einstein College of Medicine / Montefiore Medical Center; Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine / Montefiore Medical Center; Gruss-Lipper Biophotonics Center, Einstein College of Medicine / Montefiore Medical Center;
| |
Collapse
|
38
|
Abstract
The phenomenon of swarming has long been observed in nature as a strategic event that serves as a good offense toward prey and predators. Imaging studies have uncovered that neutrophils employ this swarm-like tactic within infected and inflamed tissues as part of the innate immune response. Much of our understanding of neutrophil swarming builds from observations during sterile inflammation and various bacterial, fungal, and parasitic infections of the skin. However, the architecture and function of the skin differ significantly from vital organs where highly specialized microenvironments carry out critical functions. Therefore, the detrimental extent this perturbation may have on organ function remains unclear. In this review, we examine organ-specific swarming within the skin, liver, and lungs, with a detailed focus on swarming within microvascular environments. In addition, we examine potential "swarmulants" that initiate both transient and persistent swarms that have been implicated in disease.
Collapse
Affiliation(s)
- Luke Brown
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Bryan G. Yipp
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
39
|
Pessôa-Pereira D, Scorza BM, Cyndari KI, Beasley EA, Petersen CA. Modulation of Macrophage Redox and Apoptotic Processes to Leishmania infantum during Coinfection with the Tick-Borne Bacteria Borrelia burgdorferi. Pathogens 2023; 12:1128. [PMID: 37764937 PMCID: PMC10537792 DOI: 10.3390/pathogens12091128] [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: 07/28/2023] [Revised: 09/01/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Canine leishmaniosis (CanL) is a zoonotic disease caused by protozoan Leishmania infantum. Dogs with CanL are often coinfected with tick-borne bacterial pathogens, including Borrelia burgdorferi in the United States. These coinfections have been causally associated with hastened disease progression and mortality. However, the specific cellular mechanisms of how coinfections affect microbicidal responses against L. infantum are unknown. We hypothesized that B. burgdorferi coinfection impacts host macrophage effector functions, prompting L. infantum intracellular survival. In vitro experiments demonstrated that exposure to B. burgdorferi spirochetes significantly increased L. infantum parasite burden and pro-inflammatory responses in DH82 canine macrophage cells. Induction of cell death and generation of mitochondrial ROS were significantly decreased in coinfected DH82 cells compared to uninfected and L. infantum-infected cells. Ex vivo stimulation of PBMCs from L. infantum-seronegative and -seropositive subclinical dogs with spirochetes and/or total Leishmania antigens promoted limited induction of IFNγ. Coexposure significantly induced expression of pro-inflammatory cytokines and chemokines associated with Th17 differentiation and neutrophilic and monocytic recruitment in PBMCs from L. infantum-seropositive dogs. Excessive pro-inflammatory responses have previously been shown to cause CanL pathology. This work supports effective tick prevention and risk management of coinfections as critical strategies to prevent and control L. infantum progression in dogs.
Collapse
Affiliation(s)
- Danielle Pessôa-Pereira
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA; (D.P.-P.); (B.M.S.); (E.A.B.)
- Center for Emerging Infectious Diseases, University of Iowa, Iowa City, IA 52242, USA;
| | - Breanna M. Scorza
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA; (D.P.-P.); (B.M.S.); (E.A.B.)
- Center for Emerging Infectious Diseases, University of Iowa, Iowa City, IA 52242, USA;
| | - Karen I. Cyndari
- Center for Emerging Infectious Diseases, University of Iowa, Iowa City, IA 52242, USA;
- Department of Emergency Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Erin A. Beasley
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA; (D.P.-P.); (B.M.S.); (E.A.B.)
- Center for Emerging Infectious Diseases, University of Iowa, Iowa City, IA 52242, USA;
| | - Christine A. Petersen
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA; (D.P.-P.); (B.M.S.); (E.A.B.)
- Center for Emerging Infectious Diseases, University of Iowa, Iowa City, IA 52242, USA;
| |
Collapse
|
40
|
Baars I, Jaedtka M, Dewitz LA, Fu Y, Franz T, Mohr J, Gintschel P, Berlin H, Degen A, Freier S, Rygol S, Schraven B, Kahlfuß S, van Zandbergen G, Müller AJ. Leishmania major drives host phagocyte death and cell-to-cell transfer depending on intracellular pathogen proliferation rate. JCI Insight 2023; 8:e169020. [PMID: 37310793 PMCID: PMC10443809 DOI: 10.1172/jci.insight.169020] [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: 01/19/2023] [Accepted: 06/05/2023] [Indexed: 06/15/2023] Open
Abstract
The virulence of intracellular pathogens relies largely on the ability to survive and replicate within phagocytes but also on release and transfer into new host cells. Such cell-to-cell transfer could represent a target for counteracting microbial pathogenesis. However, our understanding of the underlying cellular and molecular processes remains woefully insufficient. Using intravital 2-photon microscopy of caspase-3 activation in the Leishmania major-infected (L. major-infected) live skin, we showed increased apoptosis in cells infected by the parasite. Also, transfer of the parasite to new host cells occurred directly without a detectable extracellular state and was associated with concomitant uptake of cellular material from the original host cell. These in vivo findings were fully recapitulated in infections of isolated human phagocytes. Furthermore, we observed that high pathogen proliferation increased cell death in infected cells, and long-term residency within an infected host cell was only possible for slowly proliferating parasites. Our results therefore suggest that L. major drives its own dissemination to new phagocytes by inducing host cell death in a proliferation-dependent manner.
Collapse
Affiliation(s)
- Iris Baars
- Experimental Immunodynamics, Institute of Molecular and Clinical Immunology, Medical Faculty, and
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Moritz Jaedtka
- Division of Immunology, Paul Ehrlich Institute, Langen, Germany
- Institute for Immunology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Leon-Alexander Dewitz
- Experimental Immunodynamics, Institute of Molecular and Clinical Immunology, Medical Faculty, and
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Yan Fu
- Experimental Immunodynamics, Institute of Molecular and Clinical Immunology, Medical Faculty, and
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Tobias Franz
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Juliane Mohr
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Patricia Gintschel
- Experimental Immunodynamics, Institute of Molecular and Clinical Immunology, Medical Faculty, and
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Hannes Berlin
- Experimental Immunodynamics, Institute of Molecular and Clinical Immunology, Medical Faculty, and
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Angelina Degen
- Experimental Immunodynamics, Institute of Molecular and Clinical Immunology, Medical Faculty, and
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Sandra Freier
- Experimental Immunodynamics, Institute of Molecular and Clinical Immunology, Medical Faculty, and
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Stefan Rygol
- Experimental Immunodynamics, Institute of Molecular and Clinical Immunology, Medical Faculty, and
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Burkhart Schraven
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Sascha Kahlfuß
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Ger van Zandbergen
- Division of Immunology, Paul Ehrlich Institute, Langen, Germany
- Institute for Immunology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Andreas J. Müller
- Experimental Immunodynamics, Institute of Molecular and Clinical Immunology, Medical Faculty, and
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
41
|
Costa CHN, Chang KP, Costa DL, Cunha FVM. From Infection to Death: An Overview of the Pathogenesis of Visceral Leishmaniasis. Pathogens 2023; 12:969. [PMID: 37513817 PMCID: PMC10384967 DOI: 10.3390/pathogens12070969] [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: 05/13/2023] [Revised: 07/02/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Kala-azar, also known as visceral leishmaniasis (VL), is a disease caused by Leishmania infantum and L. donovani. Patients experience symptoms such as fever, weight loss, paleness, and enlarged liver and spleen. The disease also affects immunosuppressed individuals and has an overall mortality rate of up to 10%. This overview explores the literature on the pathogenesis of preclinical and clinical stages, including studies in vitro and in animal models, as well as complications and death. Asymptomatic infection can result in long-lasting immunity. VL develops in a minority of infected individuals when parasites overcome host defenses and multiply in tissues such as the spleen, liver, and bone marrow. Hepatosplenomegaly occurs due to hyperplasia, resulting from parasite proliferation. A systemic inflammation mediated by cytokines develops, triggering acute phase reactants from the liver. These cytokines can reach the brain, causing fever, cachexia and vomiting. Similar to sepsis, disseminated intravascular coagulation (DIC) occurs due to tissue factor overexpression. Anemia, hypergammaglobulinemia, and edema result from the acute phase response. A regulatory response and lymphocyte depletion increase the risk of bacterial superinfections, which, combined with DIC, are thought to cause death. Our understanding of VL's pathogenesis is limited, and further research is needed to elucidate the preclinical events and clinical manifestations in humans.
Collapse
Affiliation(s)
- Carlos H N Costa
- Centro de Investigações em Agravos Tropicais Emergentes e Negligenciados, Instituto de Doenças Tropicais Natan Portella, Universidade Federal do Piauí, Rua Artur de Vasconcelos 151-Sul, Teresina 64002-510, PI, Brazil
| | - Kwang-Poo Chang
- Department of Microbiology/Immunology, Center for Cancer Cell Biology, Immunology & Infection, Chicago Medical School, Rosalind Franklin University, North Chicago, IL 60064, USA
| | - Dorcas L Costa
- Centro de Investigações em Agravos Tropicais Emergentes e Negligenciados, Instituto de Doenças Tropicais Natan Portella, Universidade Federal do Piauí, Rua Artur de Vasconcelos 151-Sul, Teresina 64002-510, PI, Brazil
| | - Francisco Valmor M Cunha
- Departament of Physiotherapy, Centro Universitário Uninovafapi, Rua Vitorino Orthiges Fernandes, 6123-Uruguai, Teresina 64073-505, PI, Brazil
| |
Collapse
|
42
|
Sequera HDG, de Souza JL, Junior JDES, da Silva LS, Pinheiro SK, Kerr HKA, de Souza MLG, Guerra MVDF, de Mesquita TGR, Ramasawmy R. Variants of CARD8 in Leishmania guyanensis-cutaneous leishmaniasis and influence of the variants genotypes on circulating plasma cytokines IL-1β, TNFα and IL-8. PLoS Negl Trop Dis 2023; 17:e0011416. [PMID: 37276232 PMCID: PMC10270566 DOI: 10.1371/journal.pntd.0011416] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 06/15/2023] [Accepted: 05/24/2023] [Indexed: 06/07/2023] Open
Abstract
Nucleotide-binding oligomerization domain, leucine-rich repeat-containing protein family (NLR) are intracellular pathogen recognition receptors mediating innate immunity, releasing proinflammatory cytokines IL-1β and IL-18, and promoting pyroptotic cell death, upon sensing pathogenic or endogenous danger signals. In animal models, NLRP3 inflammasome has a dual role, pathogenic or protective in Leishmania-infection, depending on the Leishmania species and mice strain. Caspase recruitment containing domain 8 (CARD8) is a negative regulator of NLRP3 inflammasome and also an inhibitor of transcription factor NFĸB, a major transcription factor of proinflammatory cytokines. We investigated whether single nucleotide variants in CARD8 may partially explain why only a proportion of individuals coming from the same area of endemicity of leishmaniasis develop cutaneous leishmaniasis caused by Leishmania guyanensis. We genotyped four single nucleotide variants of the CARD8 gene by direct nucleotide sequencing in 1741 individuals from an endemic area of leishmaniasis, constituting 850 patients with CL and 891 healthy controls. The frequencies of the genotypes of the variants rs2288877 T>C, rs73944113 C>T, and rs2043211 A>T are similar among the patients with CL and HC, while the variant rs2288876 A>G) reveals an excess of the genotype AA among the patients with CL (44%) compared to 37% in the HC group. Allele A of the variant rs2288876 A>G) is associated with susceptibility to CL (OR = 1.2 [95%CI 1.03-1.4]; P = 0.01). Haplotype analysis showed that individuals harboring the haplotype CCAA have 280% odds of developing CL caused by L. guyanensis (OR = 3.8 [95% CI 2.0-7.7]; p = 0.00004). The variants rs2288877 T>C and rs2288876 A>G correlate with the plasma level of IL-8. Spearman correlation showed a significant positive correlation between the rs2288876 A>G allele A and the level of IL-8 (ρ = 0.22; p = 0.0002). CARD8 may partially contribute to the development of CL caused by L. guyanensis.
Collapse
Affiliation(s)
| | - Josué Lacerda de Souza
- Faculdade de Medicina Nilton Lins, Universidade Nilton Lins, Manaus, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas, Manaus, Brazil
| | - José do Espírito Santo Junior
- Faculdade de Medicina Nilton Lins, Universidade Nilton Lins, Manaus, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas, Manaus, Brazil
| | - Lener Santos da Silva
- Faculdade de Medicina Nilton Lins, Universidade Nilton Lins, Manaus, Brazil
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia da Amazonia Legal (Rede Bionorte), Universidade do Estado do Amazonas, Manaus, Brazil
| | - Suzana Kanawati Pinheiro
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Brazil
- Faculdade de Medicina Nilton Lins, Universidade Nilton Lins, Manaus, Brazil
| | | | | | - Marcus Vinitius de Farias Guerra
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Brazil
- Fundação de Medicina Tropical Doutor Heitor Vieira Dourado, Manaus, Brazil
| | - Tirza Gabrielle Ramos de Mesquita
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Brazil
- Fundação de Medicina Tropical Doutor Heitor Vieira Dourado, Manaus, Brazil
| | - Rajendranath Ramasawmy
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Brazil
- Faculdade de Medicina Nilton Lins, Universidade Nilton Lins, Manaus, Brazil
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia da Amazonia Legal (Rede Bionorte), Universidade do Estado do Amazonas, Manaus, Brazil
- Fundação de Medicina Tropical Doutor Heitor Vieira Dourado, Manaus, Brazil
- Genomic Health Surveillance Network: Optimization of Assistance and Research in the State of Amazonas (REGESAM), Manaus, Brazil
| |
Collapse
|
43
|
Valadares DG, Clay OS, Chen Y, Scorza BM, Cassel SL, Sutterwala FS, Wilson ME. NLRP12-expressing dendritic cells mediate both dissemination of infection and adaptive immune responses in visceral leishmaniasis. iScience 2023; 26:106163. [PMID: 36879824 PMCID: PMC9985045 DOI: 10.1016/j.isci.2023.106163] [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: 07/07/2022] [Revised: 10/26/2022] [Accepted: 01/02/2023] [Indexed: 02/10/2023] Open
Abstract
The NLR protein NLRP12 contributes to innate immunity, but the mechanism remains elusive. Infection of Nlrp12 -/- or wild-type mice with Leishmania infantum led to aberrant parasite tropism. Parasites replicated to higher levels in livers of Nlrp12 -/- mice than in the livers of WT mice and failed to disseminate to spleens. Most retained liver parasites resided in dendritic cells (DCs), with correspondingly fewer infected DCs in spleens. Furthermore, Nlrp12 -/- DCs expressed lower CCR7 than WT DCs, failed to migrate toward CCL19 or CCL21 in chemotaxis assays, and migrated poorly to draining lymph nodes after sterile inflammation. Leishmania-infected Nlpr12 -/- DCs were significantly less effective at transporting parasites to lymph nodes than WT DCs. Consistently, adaptive immune responses were also impaired in infected Nlrp12 -/- mice. We hypothesize that Nlrp12-expressing DCs are required for efficient dissemination and immune clearance of L. infantum from the site of initial infection. This is at least partly due to the defective expression of CCR7.
Collapse
Affiliation(s)
- Diogo Garcia Valadares
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Owen Scott Clay
- Department of Pediatrics, Division of Pediatric Rheumatology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Yani Chen
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Breanna Mary Scorza
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA
| | - Suzanne Louise Cassel
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Fayyaz Shiraz Sutterwala
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Mary Edythe Wilson
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA
- Veterans’ Affairs Medical Center, Iowa City, IA 52246, USA
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
44
|
de Korne CM, van Lieshout L, van Leeuwen FWB, Roestenberg M. Imaging as a (pre)clinical tool in parasitology. Trends Parasitol 2023; 39:212-226. [PMID: 36641293 DOI: 10.1016/j.pt.2022.12.008] [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: 11/17/2022] [Revised: 12/23/2022] [Accepted: 12/23/2022] [Indexed: 01/13/2023]
Abstract
Imaging of parasites is central to diagnosis of many parasitic diseases and has thus far played an important role in the development of antiparasitic strategies. The development of novel imaging technologies has revolutionized medicine in fields other than parasitology and has also opened up new avenues for the visualization of parasites. Here we review the role imaging technology has played so far in parasitology and how it may spur further advancement. We point out possibilities to improve current microscopy-based diagnostic methods and how to extend them with radiological imaging modalities. We also highlight in vivo tracking of parasites as a readout for efficacy of new antiparasitic strategies and as a source of fundamental insights for rational design.
Collapse
Affiliation(s)
- Clarize Maria de Korne
- Leiden University Center for Infectious Diseases, Leiden University Medical Centre, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands; Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands
| | - Lisette van Lieshout
- Leiden University Center for Infectious Diseases, Leiden University Medical Centre, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands
| | - Fijs Willem Bernhard van Leeuwen
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands
| | - Meta Roestenberg
- Leiden University Center for Infectious Diseases, Leiden University Medical Centre, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands.
| |
Collapse
|
45
|
de Sá KSG, Amaral LA, Rodrigues TS, Ishimoto AY, de Andrade WAC, de Almeida L, Freitas-Castro F, Batah SS, Oliveira SC, Pastorello MT, Fabro AT, Zamboni DS. Gasdermin-D activation promotes NLRP3 activation and host resistance to Leishmania infection. Nat Commun 2023; 14:1049. [PMID: 36828815 PMCID: PMC9958042 DOI: 10.1038/s41467-023-36626-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/09/2023] [Indexed: 02/26/2023] Open
Abstract
Intracellular parasites from the Leishmania genus cause Leishmaniasis, a disease affecting millions of people worldwide. NLRP3 inflammasome is key for disease outcome, but the molecular mechanisms upstream of the inflammasome activation are still unclear. Here, we demonstrate that despite the absence of pyroptosis, Gasdermin-D (GSDMD) is active at the early stages of Leishmania infection in macrophages, allowing transient cell permeabilization, potassium efflux, and NLRP3 inflammasome activation. Further, GSDMD is processed into a non-canonical 25 kDa fragment. Gsdmd-/- macrophages and mice exhibit less NLRP3 inflammasome activation and are highly susceptible to infection by several Leishmania species, confirming the role of GSDMD for inflammasome-mediated host resistance. Active NLRP3 inflammasome and GSDMD are present in skin biopsies of patients, demonstrating activation of this pathway in human leishmaniasis. Altogether, our findings reveal that Leishmania subverts the normal functions of GSDMD, an important molecule to promote inflammasome activation and immunity in Leishmaniasis.
Collapse
Affiliation(s)
- Keyla S G de Sá
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Luana A Amaral
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Tamara S Rodrigues
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Adriene Y Ishimoto
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Warrison A C de Andrade
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Leticia de Almeida
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Felipe Freitas-Castro
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Sabrina S Batah
- Departamento de Patologia e Medicina Legal, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Sergio C Oliveira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Mônica T Pastorello
- Departamento de Patologia e Medicina Legal, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Alexandre T Fabro
- Departamento de Patologia e Medicina Legal, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
- Serviço de Patologia do Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Dario S Zamboni
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil.
| |
Collapse
|
46
|
Sánchez-García L, Pérez-Torres A, Gudiño-Zayas ME, Zamora-Chimal J, Meneses C, Kamhawi S, Valenzuela JG, Becker I. Leishmania major-Infected Phlebotomus duboscqi Sand Fly Bites Enhance Mast Cell Degranulation. Pathogens 2023; 12:207. [PMID: 36839479 PMCID: PMC9960273 DOI: 10.3390/pathogens12020207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/31/2022] [Accepted: 01/16/2023] [Indexed: 02/01/2023] Open
Abstract
Leishmania parasites infect mammalian hosts through the bites of sand fly vectors. The response by mast cells (MC) to the parasite and vector-derived factors, delivered by sand fly bites, has not been characterized. We analyzed MC numbers and their mediators in BALB/c mice naturally infected in the ear with Leishmania major through the bite of the sand fly vector Phlebotomus duboscqi and compared them to non-infected sand fly bites. MC were found at the bite sites of infective and non-infected sand flies throughout 48 h, showing the release of granules with intense TNF-α, histamine, and tryptase staining. At 30 min and 48 h, the MC numbers were significantly higher (p < 0.001) in infected as compared to non-infected bites or controls. Neutrophil recruitment was intense during the first 6 h in the skin of infected and non-infected sand fly bites and decreased thereafter. An influx of neutrophils also occurred in lymph nodes, where a strong TNF-α stain was observed in mononuclear cells. Our data show that MC orchestrate an early inflammatory response after infected and non-infected sand fly bites, leading to neutrophilic recruitment, which potentially provides a safe passage for the parasite within the mammalian host.
Collapse
Affiliation(s)
- Laura Sánchez-García
- División Ciencias de la Salud, Universidad Autónoma del Estado de Quintana Roo, Chetumal C.P. 77039, Mexico
| | - Armando Pérez-Torres
- Departamento de Biología Celular y Tisular, Laboratorio de Inmunología Comparada de Piel y Mucosas, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Ciudad de México C.P. 04510, Mexico
| | - Marco E. Gudiño-Zayas
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Ciudad de México C.P. 04510, Mexico
| | - Jaime Zamora-Chimal
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Ciudad de México C.P. 04510, Mexico
| | - Claudio Meneses
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Jesus G. Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Ingeborg Becker
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Ciudad de México C.P. 04510, Mexico
| |
Collapse
|
47
|
Osorio EY, Uscanga-Palomeque A, Patterson GT, Cordova E, Travi BL, Soong L, Melby PC. Malnutrition-related parasite dissemination from the skin in visceral leishmaniasis is driven by PGE2-mediated amplification of CCR7-related trafficking of infected inflammatory monocytes. PLoS Negl Trop Dis 2023; 17:e0011040. [PMID: 36630476 PMCID: PMC9873180 DOI: 10.1371/journal.pntd.0011040] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 01/24/2023] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
People are infected with Leishmania donovani when the parasite is deposited in the dermis during the blood meal of the sand fly vector. Most infected people develop a subclinical latent infection, but some develop progressive visceral leishmaniasis. Malnutrition is a risk factor for the development of active VL. We previously demonstrated increased parasite dissemination from the skin to visceral organs in a murine model of malnutrition. Here we investigated the mechanism of early parasite dissemination. After delivery of L. donovani to the skin, we found enhanced capture of parasites by inflammatory monocytes and neutrophils in the skin of malnourished mice. However, parasite dissemination in malnourished mice was driven primarily by infected inflammatory monocytes, which showed increased CCR7 expression, greater intrinsic migratory capacity, and increased trafficking from skin to spleen. PGE2 production, which was increased at the site of skin infection, increased monocyte CCR7 expression and promoted CCR7-related monocyte-mediated early parasite dissemination in malnourished mice. Parasite dissemination in monocytes was reduced by inhibition of PGE2, knockdown or silencing of CCR7 in monocytes, and depletion of inflammatory monocytes through administration of diphtheria toxin to CSFR1-DTR transgenic mice that have monocyte-specific DT receptor expression. CCR7-driven trafficking of infected inflammatory monocytes through the lymph node was accompanied by increased expression of its ligands CCL19 and CCL21. These results show that the CCR7/PGE2 axis is responsible for the increased trafficking of L. donovani-infected inflammatory monocytes from the skin to the spleen in the malnourished host. Undernutrition and production of PGE2 are potential targets to reduce the risk of people developing VL. Nutritional interventions that target improved immune function and reduced PGE2 synthesis should be studied in people at risk of developing VL.
Collapse
Affiliation(s)
- E. Yaneth Osorio
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail: (PCM); (EYO)
| | - Ashanti Uscanga-Palomeque
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Grace T. Patterson
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Erika Cordova
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Bruno L. Travi
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Tropical Diseases and Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Tropical Diseases and Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Peter C. Melby
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Tropical Diseases and Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail: (PCM); (EYO)
| |
Collapse
|
48
|
Infection and Immunity. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00007-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
49
|
Carriere E, Abdul Hamid AI, Feki I, Dubuffet A, Delbac F, Gueirard P. A mouse ear skin model to study the dynamics of innate immune responses against the microsporidian Encephalitozoon cuniculi. Front Microbiol 2023; 14:1168970. [PMID: 37125152 PMCID: PMC10136781 DOI: 10.3389/fmicb.2023.1168970] [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: 02/18/2023] [Accepted: 03/29/2023] [Indexed: 05/02/2023] Open
Abstract
Microsporidia are obligate intracellular parasites related to fungi that cause severe infections in immunocompromised individuals. Encephalitozoon cuniculi is a microsporidian species capable of infecting mammals, including human and rodents. In response to microsporidian infection, innate immune system serves as the first line of defense and allows a partial clearance of the parasite via the innate immune cells, namely macrophages, neutrophils, dendritic cells, and Natural Killer cells. According to the literature, microsporidia bypass this response in vitro by modulating the response of macrophages. In order to study host-parasites interactions in vivo, we developed a model using the mouse ear pinna in combination with an intravital imaging approach. Fluorescent E. cuniculi spores were inoculated into the skin tissue to follow for the first time in real time in an in vivo model the recruitment dynamics of EGFP + phagocytic cells in response to the parasite. The results show that parasites induce an important inflammatory recruitment of phagocytes, with alterations of their motility properties (speed, displacement length, straightness). This cellular response persists in the injection zone, with spores detected inside the phagocytes up to 72 h post-infection. Immunostainings performed on ear tissue cryosections evoke the presence of developing infectious foci from 5 days post-infection, in favor of parasite proliferation in this tissue. Overall, the newly set up mice ear pinna model will increase our understanding of the immunobiology of microsporidia and in particular, to know how they can bypass and hijack the host immune system of an immunocompetent or immunosuppressed host.
Collapse
|
50
|
Omics Approaches in Drug Development against Leishmaniasis: Current Scenario and Future Prospects. Pathogens 2022; 12:pathogens12010039. [PMID: 36678387 PMCID: PMC9866966 DOI: 10.3390/pathogens12010039] [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: 12/01/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/28/2022] Open
Abstract
Leishmaniasis is a zoonotic disease transmitted in humans by the bite of Leishmania-infected phlebotomine sandflies. Each year approximately 58,500 cases of leishmaniasis are diagnosed across the globe, with a mortality rate of nearly seven percent. There are over 20 parasitic strains of Leishmania which are known to cause distinct types of leishmaniasis and pose an endemic threat to humans worldwide. Therefore, it is crucial to develop potential medications and vaccines to combat leishmaniasis. However, the task of developing therapeutic solutions is challenging due to Leishmania's digenetic lifecycle. The challenge is further intensified by cases of resistance against the available drugs. Owing to these challenges, the conventional drug development regimen is further limited by target discovery and ligand suitability for the targets. On the other hand, as an added advantage, the emergence of omics-based tools, such as high-end proteomics, transcriptomics and genomics, has hastened the pace of target discovery and target-based drug development. It is now becoming apparent that multi-omics convergence and an inter-connected systems approach is less time-consuming and more cost-effective for any drug-development process. This comprehensive review is an attempt to summarize the current knowledge on the muti-omics approach in drug development against leishmaniasis. In particular, it elaborates the potential target identification from secreted proteins in various stages of Leishmania infection and also illustrates the convergence of transcriptomic and genomic data towards the collective goal of drug discovery. This review also provides an understanding of the potential parasite's drug targets and drug resistance characteristics of the parasite, which can be used in designing effective and specific therapeutics.
Collapse
|