1
|
Roy M, Bagchi A, Karmakar C, Chatterjee M. Transdifferentiation of neutrophils facilitates the establishment of infection by Leishmania donovani parasites. Infect Immun 2025:e0040924. [PMID: 40340446 DOI: 10.1128/iai.00409-24] [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: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 05/10/2025] Open
Abstract
Neutrophil transdifferentiation involves the acquisition of dendritic cell-like properties, challenging the traditional view of neutrophils being solely phagocytes. The presence of transdifferentiated neutrophils is established in Visceral Leishmaniasis, but not in its dermal sequel, Post Kala-azar Dermal Leishmaniasis. Accordingly, this study investigated the altered functionalities of neutrophils focusing on the acquisition of dendritic cell-like properties and its impact on infection establishment. In PKDL cases, immunophenotyping of neutrophil-dendritic cells (N-DC hybrids) was performed using flow cytometry, along with studying the status of N-DC hybrid inducing cytokines (TNF-α, IFN-γ) and growth factor (GM-CSF). Ex vivo infection of neutrophils with L. donovani was monitored by droplet digital PCR, employing A2; additionally, their frequency of transdifferentiation, oxidative and phagocytic status, as well as apoptosis potential were quantified by flow cytometry. Compared with healthy controls, neutrophils from PKDL cases demonstrated a significant upregulation of CD83 positivity, but the frequency of co-stimulation (HLA-DR, CD80/86) was unaltered. PKDL cases demonstrated raised levels of TNF-α and IFN-γ, but GM-CSF remained unchanged. Following ex vivo infection of neutrophils, infection was evident at 2 h and was accompanied by CD83 positivity. Furthermore, the CD66b+/CD83 vis-à-vis CD66b+/CD83- subset exhibited heightened generation of reactive oxygen species (ROS), enhanced phagocytosis, and increased apoptosis. Taken together, neutrophils from PKDL cases demonstrated transdifferentiation with the absence of antigen-presenting function. Virulent Leishmania induced transdifferentiation in neutrophils, altering their functionalities and facilitating parasite uptake, along with heightened generation of intra-neutrophilic ROS and enhanced apoptosis, which possibly facilitated their engulfment by macrophages, thereby bolstering the "Trojan horse" mechanism of parasite transfer.
Collapse
Affiliation(s)
- Madhurima Roy
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research (IPGME&R), Kolkata, India
| | - Aniruddha Bagchi
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research (IPGME&R), Kolkata, India
| | - Chaitali Karmakar
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research (IPGME&R), Kolkata, India
| | - Mitali Chatterjee
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research (IPGME&R), Kolkata, India
| |
Collapse
|
2
|
Roy M, Sengupta R, Chakraborty BC, Chatterjee U, von Stebut E, Kaye PM, Chatterjee M. Role of neutrophils in the pathogenesis of Post Kala-azar Dermal Leishmaniasis (PKDL). PLoS Negl Trop Dis 2024; 18:e0012655. [PMID: 39602398 PMCID: PMC11602034 DOI: 10.1371/journal.pntd.0012655] [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: 04/26/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Post Kala-azar Dermal Leishmaniasis (PKDL) is a dermal sequel of visceral leishmaniasis (VL), poses a significant threat to the success of ongoing kala-azar elimination program, due to its potential role in sustaining transmission cycles and complicating disease management strategies. In VL, neutrophils have been identified as the 'first line of defence', having multiple roles in disease pathogenesis, but their role in PKDL, if any, still remains elusive; presenting a critical gap in knowledge, and was the aim of this study. METHODOLOGY/PRINCIPAL FINDINGS In a cohort of PKDL patients, CD66b+ neutrophils were quantified in skin biopsies, followed by immunostaining of FFPE sections to identify activated neutrophils (CD66b+/CD64+) and degranulated (CD66b+/MPO+), along with expression of neutrophil elastase (NE), matrix metalloprotease 9 (MMP9) and collagen I. Plasma levels of neutrophil chemo-attractants CXCL8/1/2/5, CCL2 and 20 and cytokines, (IL-6, IFN-γ, IL-4, IL-10, TNF-α, IL-17 and IL-22, 23) were evaluated by a multiplex assay, while lesional expression of IL-8, IL-10 and IL-17 was evaluated by immunohistochemistry. As compared to healthy individuals (control skin samples), PKDL cases at the lesional sites had an increased number of activated CD66b+ neutrophils (positive for CD64+, MPO+ and NE+). The plasma levels of neutrophil chemo-attractants, pro-inflammatory and regulatory cytokines were raised as was circulating and lesional IL-8, along with an enhanced lesional expression of IL-10 and IL-17A. An increase in circulatory and lesional MMP9 was accompanied by decreased collagen I, suggesting disintegration of matrix integrity. CONCLUSIONS/SIGNIFICANCE Taken together, in PKDL, activated neutrophils possibly contribute towards modulating the lesional landscape. Understanding this involvement of neutrophils in patients with PKDL, particularly in the absence of an animal model, could offer better understanding of the disease pathogenesis and provide insights into novel therapeutic strategies for the ongoing elimination program.
Collapse
Affiliation(s)
- Madhurima Roy
- Dept. of Pharmacology, Institute of Post Graduate Medical Education and Research (IPGME&R), Kolkata, India
| | - Ritika Sengupta
- Dept. of Pharmacology, Institute of Post Graduate Medical Education and Research (IPGME&R), Kolkata, India
| | - Bidhan Chandra Chakraborty
- Multidisciplinary Research Unit (MRU) Institute of Post Graduate Medical Education and Research (IPGME&R), Kolkata, India
| | - Uttara Chatterjee
- Pathology, Institute of Post Graduate Medical Education and Research (IPGME&R), Kolkata, India
| | - Esther von Stebut
- Department of Dermatology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Paul M. Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Mitali Chatterjee
- Dept. of Pharmacology, Institute of Post Graduate Medical Education and Research (IPGME&R), Kolkata, India
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Blasi-Brugué C, Martínez-Flórez I, Baxarias M, del Rio-Velasco J, Solano-Gallego L. Exploring the Relationship between Neutrophil Activation and Different States of Canine L. infantum Infection: Nitroblue Tetrazolium Test and IFN-γ. Vet Sci 2023; 10:572. [PMID: 37756094 PMCID: PMC10535614 DOI: 10.3390/vetsci10090572] [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: 07/19/2023] [Revised: 09/04/2023] [Accepted: 09/11/2023] [Indexed: 09/28/2023] Open
Abstract
This study aimed to investigate the role of neutrophils in canine leishmaniosis by assessing neutrophil activation and its relationship with different states of L. infantum infection and antibody and IFN-γ production. Dogs were categorized into five groups: healthy-seronegative (n = 25), healthy-seropositive (n = 21), LeishVet-stage I (n = 25), Leishvet-stage II (n = 41), and LeishVet-stage III-IV (n = 16). Results of the nitroblue tetrazolium reduction test (NBT) showed significantly higher neutrophil activation in stage I (median:17.17, range: [7.33-31.50]%) compared to in healthy-seronegative (4.10 [1.20-18.00]%), healthy-seropositive (7.65 [3.98-21.74]%), stage II (6.50 [1.50-28.70]%), and stage III-IV (7.50 [3.00-16.75]%) groups (p < 0.0001). Healthy-seropositive dogs also displayed higher values than all groups except stage I. Stages II and III-IV did not show significant differences compared to healthy-seronegative. Regarding IFN-γ, stage I dogs had higher concentrations (median:127.90, range: [0-3998.00] pg/mL) than healthy-seronegative (0 [0-109.50] pg/mL) (p = 0.0002), stage II (9.00 [0-5086.00] pg/mL) (p = 0.045), and stage III-IV (3.50 [80.00-548.80] pg/mL) (p = 0.02) dogs. Stage II dogs showed increased IFN-γ compared to healthy-seronegative dogs (p = 0.015), while stage III-IV dogs had no significant differences compared to healthy-seronegative dogs (p = 0.12). Healthy-seropositive dogs had elevated IFN-γ concentrations compared to healthy-seronegative dogs (p = 0.001) and dogs in stage III-IV (p = 0.03). In conclusion, neutrophil activation was higher in dogs with mild disease and healthy-seropositive dogs, and a relationship between neutrophil activation and the production of IFN-γ was found.
Collapse
Affiliation(s)
| | | | | | | | - Laia Solano-Gallego
- Departament de Medicina i Cirurgia Animals, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; (C.B.-B.); (I.M.-F.); (M.B.); (J.d.R.-V.)
| |
Collapse
|
6
|
Zhou Z, Yang W, Yu T, Yu Y, Zhao X, Yu Y, Gu C, Bilotta AJ, Yao S, Zhao Q, Golovko G, Li M, Cong Y. GPR120 promotes neutrophil control of intestinal bacterial infection. Gut Microbes 2023; 15:2190311. [PMID: 36927391 PMCID: PMC10026904 DOI: 10.1080/19490976.2023.2190311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
G-protein coupled receptor 120 (GPR 120) has been implicated in anti-inflammatory functions. However, how GPR120 regulates the neutrophil function remains unknown. This study investigated the role of GPR120 in the regulation of neutrophil function against enteric bacteria. 16S rRNA sequencing was used for measuring the gut microbiota of wild-type (WT) mice and Gpr120-/- mice. Citrobacter rodentium infection and dextran sulfate sodium (DSS)-induced colitis models were performed in WT and Gpr120-/- mice. Mouse peritoneal-derived primary neutrophils were used to determine the neutrophil functions. Gpr120-/- mice showed altered microbiota composition. Gpr120-/- mice exhibited less capacity to clear intestinal Citrobacter rodentium and more severe intestinal inflammation upon infection or DSS insults. Depletion of neutrophils decreased the intestinal clearance of Citrobacter rodentium. GPR120 agonist, CpdA, enhanced WT neutrophil production of reactive oxygen species (ROS) and extracellular traps (NETs), and GPR120-deficient neutrophils demonstrated a lower level of ROS and NETs. CpdA-treated neutrophils showed an enhanced capacity to inhibit the growth of Citrobacter rodentium, which was abrogated by the inhibition of either NETs or ROS. CpdA promoted neutrophil inhibition of the growth of commensal bacteria Escherichia coli O9:H4 and pathobiont Escherichia coli O83:H1 isolated from a Crohn's disease patient. Mechanically, mTOR activation and glycolysis mediated GPR120 induction of ROS and NETs in neutrophils. Additionally, CpdA promoted the neutrophil production of IL-17 and IL-22, and treatment with a conditioned medium of GPR120-activated neutrophils increased intestinal epithelial cell barrier functions. Our study demonstrated the critical role of GPR120 in neutrophils in protection against enteric bacterial invasion.
Collapse
Affiliation(s)
- Zheng Zhou
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, USA
- Department of Gastroenterology, Nan Fang Hospital, Southern Medical University, Guangzhou, China
- Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenjing Yang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, USA
| | - Tianming Yu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, USA
| | - Yu Yu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, USA
| | - Xiaojing Zhao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, USA
| | - Yanbo Yu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, USA
| | - Chuncai Gu
- Department of Gastroenterology, Nan Fang Hospital, Southern Medical University, Guangzhou, China
| | - Anthony J Bilotta
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, USA
| | - Suxia Yao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, USA
| | - Qihong Zhao
- Bristol-MyersSquibb, Princeton, New Jersey, USA
| | - George Golovko
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, USA
| | - Mingsong Li
- Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, USA
| |
Collapse
|
7
|
Jha B, Reverte M, Ronet C, Prevel F, Morgenthaler FD, Desponds C, Lye LF, Owens KL, Scarpellino L, Dubey LK, Sabine A, Petrova TV, Luther SA, Beverley SM, Fasel N. In and out: Leishmania metastasis by hijacking lymphatic system and migrating immune cells. Front Cell Infect Microbiol 2022; 12:941860. [PMID: 36034709 PMCID: PMC9414205 DOI: 10.3389/fcimb.2022.941860] [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: 05/11/2022] [Accepted: 07/19/2022] [Indexed: 11/22/2022] Open
Abstract
The lymphatic system plays a crucial role in mounting immune response against intracellular pathogens, and recent studies have documented its role in facilitating tumor dissemination linked largely with cancer cells. However, in mucocutaneous leishmaniasis (MCL) caused by Leishmania Viannia subgenus showing infectious metastasis and resulting in severe distant secondary lesions, the route of escape of these parasites to secondary sites has not yet been investigated in detail. Our results demonstrated that when infection was associated with inflammation and additionally exacerbated by the presence of dsRNA viral endosymbiont (LRV1), lymphatic vessels could serve as efficient routes for infected cells to egress from the primary site and colonize distant organs. We challenged this hypothesis by using the intracellular Leishmania protozoan parasites Leishmania guyanensis (Lgy) associated with or without a dsRNA viral endosymbiont, exacerbating the infection and responsible for a strong inflammatory response, and favoring metastasis of the infection. We analyzed possible cargo cells and the routes of dissemination through flow cytometry, histological analysis, and in vivo imaging in our metastatic model to show that parasites disseminated not only intracellularly but also as free extracellular parasites using migrating immune cells, lymph nodes (LNs), and lymph vessels, and followed intricate connections of draining and non-draining lymph node to finally end up in the blood and in distant skin, causing new lesions.
Collapse
Affiliation(s)
- Baijayanti Jha
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Marta Reverte
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Catherine Ronet
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Florence Prevel
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | | | - Chantal Desponds
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Lon-Fye Lye
- Department of Molecular Microbiology, School of Medicine, Washington University, St. Louis, MO, United States
| | - Katherine L. Owens
- Department of Molecular Microbiology, School of Medicine, Washington University, St. Louis, MO, United States
| | | | - Lalit Kumar Dubey
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- Centre for Microvascular Research, John Vane Science Centre, Queen Mary University of London, London, United Kingdom
| | - Amélie Sabine
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne and Centre Hospitalier Universitaire Vaudois, Epalinges, Switzerland
| | - Tatiana V. Petrova
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne and Centre Hospitalier Universitaire Vaudois, Epalinges, Switzerland
| | - Sanjiv A. Luther
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Stephen M. Beverley
- Department of Molecular Microbiology, School of Medicine, Washington University, St. Louis, MO, United States
| | - Nicolas Fasel
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- *Correspondence: Nicolas Fasel,
| |
Collapse
|
8
|
Venugopal G, Bird JT, Washam CL, Roys H, Bowlin A, Byrum SD, Weinkopff T. In vivo transcriptional analysis of mice infected with Leishmania major unveils cellular heterogeneity and altered transcriptomic profiling at single-cell resolution. PLoS Negl Trop Dis 2022; 16:e0010518. [PMID: 35789215 PMCID: PMC9286232 DOI: 10.1371/journal.pntd.0010518] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 07/15/2022] [Accepted: 05/18/2022] [Indexed: 01/02/2023] Open
Abstract
Leishmania parasites cause cutaneous leishmaniasis (CL), a disease characterized by disfiguring, ulcerative skin lesions. Both parasite and host gene expression following infection with various Leishmania species has been investigated in vitro, but global transcriptional analysis following L. major infection in vivo is lacking. Thus, we conducted a comprehensive transcriptomic profiling study combining bulk RNA sequencing (RNA-Seq) and single-cell RNA sequencing (scRNA-Seq) to identify global changes in gene expression in vivo following L. major infection. Bulk RNA-Seq analysis revealed that host immune response pathways like the antigen processing and presentation pathway were significantly enriched amongst differentially expressed genes (DEGs) upon infection, while ribosomal pathways were significantly downregulated in infected mice compared to naive controls. scRNA-Seq analyses revealed cellular heterogeneity including distinct resident and recruited cell types in the skin following murine L. major infection. Within the individual immune cell types, several DEGs indicative of many interferon induced GTPases and antigen presentation molecules were significantly enhanced in the infected ears including macrophages, resident macrophages, and inflammatory monocytes. Ingenuity Pathway Analysis of scRNA-Seq data indicated the antigen presentation pathway was increased with infection, while EIF2 signaling is the top downregulated pathway followed by eIF4/p70S6k and mTOR signaling in multiple cell types including macrophages, blood and lymphatic endothelial cells. Altogether, this transcriptomic profile highlights known recruitment of myeloid cells to lesions and recognizes a potential role for EIF2 signaling in murine L. major infection in vivo.
Collapse
Affiliation(s)
- Gopinath Venugopal
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Jordan T. Bird
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Arkansas Children’s Research Institute, Little Rock, Arkansas, United States of America
| | - Charity L. Washam
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Arkansas Children’s Research Institute, Little Rock, Arkansas, United States of America
| | - Hayden Roys
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Anne Bowlin
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Stephanie D. Byrum
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Arkansas Children’s Research Institute, Little Rock, Arkansas, United States of America
- * E-mail: (SDB); (TW)
| | - Tiffany Weinkopff
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- * E-mail: (SDB); (TW)
| |
Collapse
|
9
|
Quintela-Carvalho G, Goicochea AMC, Mançur-Santos V, Viana SDM, Luz YDS, Dias BRS, Lázaro-Souza M, Suarez M, de Oliveira CI, Saraiva EM, Brodskyn CI, Veras PT, de Menezes JP, Andrade BB, Lima JB, Descoteaux A, Borges VM. Leishmania infantum Defective in Lipophosphoglycan Biosynthesis Interferes With Activation of Human Neutrophils. Front Cell Infect Microbiol 2022; 12:788196. [PMID: 35463648 PMCID: PMC9019130 DOI: 10.3389/fcimb.2022.788196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 03/10/2022] [Indexed: 11/13/2022] Open
Abstract
Visceral leishmaniasis (VL) is often associated with hematologic manifestations that may interfere with neutrophil response. Lipophosphoglycan (LPG) is a major molecule on the surface of Leishmania promastigotes, which has been associated with several aspects of the parasite–vector–host interplay. Here, we investigated how LPG from Leishmania (L.) infantum, the principal etiological agent of VL in the New World, influences the initial establishment of infection during interaction with human neutrophils in an experimental setting in vitro. Human neutrophils obtained from peripheral blood samples were infected with either the wild-type L. infantum (WT) strain or LPG-deficient mutant (∆lpg1). In this setting, ∆lpg1 parasites displayed reduced viability compared to WT L. infantum; such finding was reverted in the complemented ∆lpg1+LPG1 parasites at 3- and 6-h post-infection. Confocal microscopy experiments indicated that this decreased survival was related to enhanced lysosomal fusion. In fact, LPG-deficient L. infantum parasites more frequently died inside neutrophil acidic compartments, a phenomenon that was reverted when host cells were treated with Wortmannin. We also observed an increase in the secretion of the neutrophil collagenase matrix metalloproteinase-8 (MMP-8) by cells infected with ∆lpg1 L. infantum compared to those that were infected with WT parasites. Furthermore, collagen I matrix degradation was found to be significantly increased in ∆lpg1 parasite-infected cells but not in WT-infected controls. Flow cytometry analysis revealed a substantial boost in production of reactive oxygen species (ROS) during infection with either WT or ∆lpg1 L. infantum. In addition, killing of ∆lpg1 parasites was shown to be more dependent on the ROS production than that of WT L. infantum. Notably, inhibition of the oxidative stress with Apocynin potentially fueled ∆lpg1 L. infantum fitness as it increased the intracellular parasite viability. Thus, our observations demonstrate that LPG may be a critical molecule fostering parasite survival in human neutrophils through a mechanism that involves cellular activation and generation of free radicals.
Collapse
Affiliation(s)
- Graziele Quintela-Carvalho
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
- Instituto Federal de Educação, Ciência e Tecnologia Baiano (IFBaiano), Alagoinhas, Brazil
| | - Astrid Madeleine Calero Goicochea
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Vanessa Mançur-Santos
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Sayonara de Melo Viana
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Yasmin da Silva Luz
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Beatriz Rocha Simões Dias
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Milena Lázaro-Souza
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Martha Suarez
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Camila Indiani de Oliveira
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Elvira M. Saraiva
- Departamento de Imunologia, Laboratório de Imunobiologia das Leishmanioses, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cláudia I. Brodskyn
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Patrícia T. Veras
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Juliana P.B. de Menezes
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Bruno B. Andrade
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
- Escola Bahiana de Medicina e Saúde Pública, Salvador, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Curso de Medicina, Faculdade de Tecnologia e Ciências, Salvador, Brazil
- Universidade Salvador (UNIFACS), Laureate Universities, Salvador, Brazil
| | - Jonilson Berlink Lima
- Núcleo de Agentes Infecciosos e Vetores (NAIVE), Universidade Federal do Oeste da Bahia (UFOB), Barreiras, Brazil
| | - Albert Descoteaux
- Institut National de la Recherche Scientifique (INRS)–Centre Armand-Frappier Santé Biotechnologie, Laval, QC, Canada
- *Correspondence: Valéria M. Borges, ; Albert Descoteaux,
| | - Valéria M. Borges
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
- *Correspondence: Valéria M. Borges, ; Albert Descoteaux,
| |
Collapse
|
10
|
Cecílio P, Cordeiro-da-Silva A, Oliveira F. Sand flies: Basic information on the vectors of leishmaniasis and their interactions with Leishmania parasites. Commun Biol 2022; 5:305. [PMID: 35379881 PMCID: PMC8979968 DOI: 10.1038/s42003-022-03240-z] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/08/2022] [Indexed: 11/09/2022] Open
Abstract
Blood-sucking arthropods transmit a variety of human pathogens acting as disseminators of the so-called vector-borne diseases. Leishmaniasis is a spectrum of diseases caused by different Leishmania species, transmitted quasi worldwide by sand flies. However, whereas many laboratories focus on the disease(s) and etiological agents, considerably less study the respective vectors. In fact, information on sand flies is neither abundant nor easy to find; aspects including basic biology, ecology, and sand-fly-Leishmania interactions are usually reported separately. Here, we compile elemental information on sand flies, in the context of leishmaniasis. We discuss the biology, distribution, and life cycle, the blood-feeding process, and the Leishmania-sand fly interactions that govern parasite transmission. Additionally, we highlight some outstanding questions that need to be answered for the complete understanding of parasite–vector–host interactions in leishmaniasis. In this review, numerous aspects of sand flies as vectors of Leishmania parasites—from biology to the vector parasite interactions—are discussed.
Collapse
Affiliation(s)
- Pedro Cecílio
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA. .,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal. .,Parasite Disease Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal. .,Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto (FFUP), Porto, Portugal.
| | - Anabela Cordeiro-da-Silva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Parasite Disease Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto (FFUP), Porto, Portugal
| | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
| |
Collapse
|
11
|
Cheng Y, Cao W, Zhang J, Wang J, Liu X, Wu Q, Lin Q. Determinants of Diabetic Peripheral Neuropathy and Their Clinical Significance: A Retrospective Cohort Study. Front Endocrinol (Lausanne) 2022; 13:934020. [PMID: 35957831 PMCID: PMC9360478 DOI: 10.3389/fendo.2022.934020] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND In this study, we investigated the epidemiological characteristics and predictors of diabetic peripheral neuropathy (DPN) in adult patients with type 2 diabetes mellitus (DM). METHODS The study was designed as a retrospective cohort trial at the First Affiliated Hospital of Wenzhou Medical University. From January 2017 to December 2020, a total of 1,262 patients with DM were enrolled to assess the risk factors for DPN. The patients were divided into two groups (DPN group and non-DPN group). The Mann-Whitney U test or t-test, receiver operating characteristic (ROC) analyses, univariate chi-square analyses, and multiple logistic regression analyses were used to analyze the adjusted predictors of DPN. RESULTS The overall prevalence of DPN in DM patients was 72.7% (n = 793/1,091). Multivariate analysis revealed that age > 66 years (odds ratio [OR], 2.647; 95% confidence interval [CI] 1.469-4.770; p = 0.002), history of hypertension (OR, 1.829; 95% CI 1.146-2.920; p = 0.011), neutrophil (NE) levels exceeding 4.0 × 109/L (OR 0.256; 95% CI 0.162-0.405; p = 0.001), lymphocyte (LY) levels over 3.0 × 109/L (OR 7.173; 95% CI 4.258-12.086; p = 0.000), HbA1c > 7.7% (OR 3.151; 95% CI 1.959-5.068; p = 0.000), and FT3 > 4.4 pmol/L (OR 0.417; 95% CI 0.263-0.662; p = 0.000) were six significant predictive factors for the prevalence of DPN. CONCLUSIONS High levels of LY, HbA1c, history of hypertension, and > 66 years of age increase the risk of DPN in adult patients with DM, while high levels of NE and FT3 were protective factors of DPN. Thus, the prediction of DPN can significantly be improved by identifying older patients over the age of 66 and history of hypertension, as well as establishing the biochemical cutoff values of NE, LY, HbA1c, and FT3.
Collapse
Affiliation(s)
- Yifan Cheng
- Department of Neurology, Center for Rehabilitation Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Wen Cao
- Department of Neurology, The Third Hospital of Peking University, Beijing, China
| | - Junzhe Zhang
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jiabin Wang
- Department of Neurology, Hebei Medical University, Shijiazhuang, China
| | - Xiang Liu
- Department of Neurology, Hebei Medical University, Shijiazhuang, China
| | - Qianqian Wu
- Department of Geriatrics, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Qingxia Lin
- Department of Psychiatry, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Qingxia Lin,
| |
Collapse
|
12
|
Gomes RS, Silva MVT, Oliveira MAP, Joosten LAB, Ribeiro-Dias F. Protective immune response mediated by neutrophils in experimental visceral leishmaniasis is enhanced by IL-32γ. Cell Immunol 2021; 371:104449. [PMID: 34784560 DOI: 10.1016/j.cellimm.2021.104449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 11/03/2022]
Abstract
Neutrophils are important cells in protection against microbial infections including visceral leishmaniasis (VL). It is well known that IL-32γ increases the protective T helper 17 cell mediated immune response against Leishmania infantum. Thus, in this study we evaluated whether IL-32 γ can increase the protective role of neutrophils against VL. In comparison with wild type (WT) mice, transgenic mice for human IL-32 γ (IL-32 γ Tg) presented a higher frequency and absolute number of neutrophils in both spleen and liver after the establishment of L. infantum infection. The IL-32 concentrations correlated with neutrophil numbers in the infected tissues. The IL-32 γ -induced recruitment of neutrophils was dependent on IL-17, since inhibition of Th17 T cells generation and IL-17 production with digoxin treatment reversed the effects of IL-32 γ. In murine neutrophils, the presence of IL-32 γ enhanced the phagocytosis of L. infantum via CR3. In addition, murine IL-32 γ Tg neutrophils were able to kill L. infantum due to the increased production of ROS when compared with WT neutrophils. In fact, IL-32 γ Tg mice lost their ability to control infection by L. infantum when neutrophils were depleted. In parallel, treatment of human neutrophils with recombinant IL-32 γ increased phagocytosis and ROS-dependent killing of L. infantum, similarly to murine IL-32 γ Tg neutrophils. The data show that IL-32 γ induces neutrophil recruitment to organs affected by VL and increases phagocytosis and killing of L. infantum by neutrophils. Together, data indicate the pivotal axis IL-32 γ -Th17-neutrophils to control VL.
Collapse
Affiliation(s)
- Rodrigo Saar Gomes
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil.
| | | | | | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Fátima Ribeiro-Dias
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil.
| |
Collapse
|
13
|
Möller S, Laskay T. Purinergic Enhancement of Anti-Leishmanial Effector Functions of Neutrophil Granulocytes. Front Immunol 2021; 12:747049. [PMID: 34733282 PMCID: PMC8558537 DOI: 10.3389/fimmu.2021.747049] [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: 07/25/2021] [Accepted: 09/06/2021] [Indexed: 11/22/2022] Open
Abstract
Although macrophages are considered for host cells for the multiplication of Leishmania, recent studies indicate the important role of neutrophil granulocytes as host cells for these intracellular parasites. Neutrophils have been shown to be massively and rapidly recruited to the site of Leishmania infection where they represent the first cells to encounter the parasites. Exposure to ATP and UTP have been shown to enhance anti-Leishmania activity of macrophages and intralesional injection of UTP led to strongly reduced parasite load in vivo. Since the in vivo anti-leishmanial effect of extracellular UTP correlated with enhanced neutrophil recruitment and enhanced ROS production at the site of Leishmania infection we hypothesized that exposure to extracellular nucleotides can directly enhance the killing of Leishmania by neutrophils. Since purinergic signaling is an essential mechanism of neutrophil activation the aim of the present study was to assess whether purinergic exposure results in the activation of anti-leishmanial neutrophil functions and, therefore, represent an essential component of enhanced anti-leishmanial defense in leishmaniasis. We could show that exposure to ATP and UTP led to activation and enhanced CD11b expression of primary human neutrophils in vitro. Leishmania-induced ROS production was strongly enhanced by extracellular ATP and UTP. Importantly, exposure to ATP and UTP resulted in enhanced killing of Leishmania donovani by neutrophils. In addition, ATP strongly enhanced the secretion of IL-8 and IL-1β by Leishmania-exposed neutrophils. Our results suggest that signaling via the P2 receptor and phosphorylation of Erk1/2, Akt and p38 are involved in the purinergic enhancement of anti-leishmanial functions of neutrophils.
Collapse
Affiliation(s)
- Sonja Möller
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Tamás Laskay
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| |
Collapse
|
14
|
Volpedo G, Pacheco-Fernandez T, Bhattacharya P, Oljuskin T, Dey R, Gannavaram S, Satoskar AR, Nakhasi HL. Determinants of Innate Immunity in Visceral Leishmaniasis and Their Implication in Vaccine Development. Front Immunol 2021; 12:748325. [PMID: 34712235 PMCID: PMC8546207 DOI: 10.3389/fimmu.2021.748325] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/24/2021] [Indexed: 12/22/2022] Open
Abstract
Leishmaniasis is endemic to the tropical and subtropical regions of the world and is transmitted by the bite of an infected sand fly. The multifaceted interactions between Leishmania, the host innate immune cells, and the adaptive immunity determine the severity of pathogenesis and disease development. Leishmania parasites establish a chronic infection by subversion and attenuation of the microbicidal functions of phagocytic innate immune cells such as neutrophils, macrophages and dendritic cells (DCs). Other innate cells such as inflammatory monocytes, mast cells and NK cells, also contribute to resistance and/or susceptibility to Leishmania infection. In addition to the cytokine/chemokine signals from the innate immune cells, recent studies identified the subtle shifts in the metabolic pathways of the innate cells that activate distinct immune signal cascades. The nexus between metabolic pathways, epigenetic reprogramming and the immune signaling cascades that drive the divergent innate immune responses, remains to be fully understood in Leishmania pathogenesis. Further, development of safe and efficacious vaccines against Leishmaniasis requires a broader understanding of the early interactions between the parasites and innate immune cells. In this review we focus on the current understanding of the specific role of innate immune cells, the metabolomic and epigenetic reprogramming and immune regulation that occurs during visceral leishmaniasis, and the strategies used by the parasite to evade and modulate host immunity. We highlight how such pathways could be exploited in the development of safe and efficacious Leishmania vaccines.
Collapse
Affiliation(s)
- Greta Volpedo
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Thalia Pacheco-Fernandez
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Parna Bhattacharya
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Timur Oljuskin
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Ranadhir Dey
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Sreenivas Gannavaram
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Abhay R Satoskar
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Hira L Nakhasi
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
15
|
Meireles PW, de Souza DPB, Rezende MG, Borsodi MPG, de Oliveira DE, da Silva LCRP, de Souza AMT, Viana GM, Rodrigues CR, do Carmo FA, de Sousa VP, Rossi-Bergmann B, Cabral LM. Nanoparticles Loaded with a New Thiourea Derivative: Development and In vitro Evaluation Against Leishmania amazonensis. Curr Drug Deliv 2021; 17:694-702. [PMID: 32621717 DOI: 10.2174/1567201817666200704132348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/27/2019] [Accepted: 02/13/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Leishmaniasis is a neglected tropical disease caused by protozoa of the genus Leishmania. Current treatments are restricted to a small number of drugs that display both severe side effects and a potential for parasites to develop resistance. A new N-(3,4-methylenedioxyphenyl)-N'- (2-phenethyl) thiourea compound (thiourea 1) has shown promising in vitro activity against Leishmania amazonensis with an IC50 of 54.14 μM for promastigotes and an IC50 of 70 μM for amastigotes. OBJECTIVE To develop a formulation of thiourea 1 as an oral treatment for leishmaniasis, it was incorporated into Nanoparticles (NPs), a proven approach to provide long-acting drug delivery systems. METHODS Poly (D,L-Lactic-co-Glycolic Acid) (PLGA) polymeric NPs containing thiourea 1 were obtained through a nanoprecipitation methodology associated with solvent evaporation. The NPs containing thiourea 1 were characterized for Encapsulation Efficiency (EE%), reaction yield (% w/w), surface charge, particle size and morphology by Transmission Electron Microscopy (TEM). RESULTS NPs with thiourea 1 showed an improved in vitro leishmanicidal activity with a reduction in its cytotoxicity against macrophages (CC50>100 μg/mL) while preserving its IC50 against intracellular amastigotes (1.46 ± 0.09 μg/mL). This represents a parasite Selectivity Index (SI) of 68.49, which is a marked advancement from the reference drug pentamidine (SI = 30.14). CONCLUSION The results suggest that the incorporation into NPs potentiated the therapeutic effect of thiourea 1, most likely by improving the selective delivery of the drug to the phagocytic cells that are targeted for infection by L. amazonensis. This work reinforces the importance of nanotechnology in the acquisition of new therapeutic alternatives for oral treatments.
Collapse
Affiliation(s)
- Paloma Wetler Meireles
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Dandara Paiva Barroso de Souza
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marianne Grilo Rezende
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | | - Gil Mendes Viana
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos Rangel Rodrigues
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Flavia Almada do Carmo
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Valeria Pereira de Sousa
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bartira Rossi-Bergmann
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lucio Mendes Cabral
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
16
|
Bulté D, Van Bockstal L, Dirkx L, Van den Kerkhof M, De Trez C, Timmermans JP, Hendrickx S, Maes L, Caljon G. Miltefosine enhances infectivity of a miltefosine-resistant Leishmania infantum strain by attenuating its innate immune recognition. PLoS Negl Trop Dis 2021; 15:e0009622. [PMID: 34292975 PMCID: PMC8330912 DOI: 10.1371/journal.pntd.0009622] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 08/03/2021] [Accepted: 07/01/2021] [Indexed: 01/08/2023] Open
Abstract
Background Miltefosine (MIL) is currently the only oral drug available to treat visceral leishmaniasis but its use as first-line monotherapy has been compromised by an increasing treatment failure. Despite the scarce number of resistant clinical isolates, MIL-resistance by mutations in a single aminophospholipid transporter gene can easily be selected in a laboratory environment. These mutations result in a reduced survival in the mammalian host, which can partially be restored by exposure to MIL, suggesting a kind of drug-dependency. Methodology/Principal findings To enable a combined study of the infection dynamics and underlying immunological events for differential in vivo survival, firefly luciferase (PpyRE9) / red fluorescent protein (DsRed) double-reporter strains were generated of MIL-resistant (MIL-R) and syngeneic MIL-sensitive (MIL-S) Leishmania infantum. Results in C57Bl/6 and BALB/c mice show that MIL-R parasites induce an increased innate immune response that is characterized by enhanced influx and infection of neutrophils, monocytes and dendritic cells in the liver and elevated serum IFN-γ levels, finally resulting in a less efficient establishment in liver macrophages. The elevated IFN-γ levels were shown to originate from an increased response of hepatic NK and NKT cells to the MIL-R parasites. In addition, we demonstrated that MIL could increase the in vivo fitness of MIL-R parasites by lowering NK and NKT cell activation, leading to a reduced IFN-γ production. Conclusions/Significance Differential induction of innate immune responses in the liver was found to underlie the attenuated phenotype of a MIL-R parasite and its peculiar feature of drug-dependency. The impact of MIL on hepatic NK and NKT activation and IFN-γ production following recognition of a MIL-R strain indicates that this mechanism may sustain infections with resistant parasites and contribute to treatment failure. Visceral leishmaniasis is a neglected tropical disease that is fatal if left untreated. Miltefosine is currently the only oral drug available but is increasingly failing to cure patients, resulting in its discontinuation as first-line drug in some endemic areas. To understand these treatment failures, we investigated the complex interplay of the parasite with the host immune system in the presence and absence of miltefosine. Our data indicate that miltefosine-resistant Leishmania parasites become severely hampered in their in vivo infectivity, which could be attributed to the induction of a pronounced innate immune response. Interestingly, the infection deficit was partially restored in the presence of miltefosine. Our results further indicate that miltefosine can exacerbate infections with resistant parasites by reducing innate immune recognition. This study provides new insights into the complex interplay between parasite, drug and host and discloses an immune-related mechanism of treatment failure.
Collapse
Affiliation(s)
- Dimitri Bulté
- University of Antwerp, Department of Biomedical Sciences, Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
| | - Lieselotte Van Bockstal
- University of Antwerp, Department of Biomedical Sciences, Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
| | - Laura Dirkx
- University of Antwerp, Department of Biomedical Sciences, Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
| | - Magali Van den Kerkhof
- University of Antwerp, Department of Biomedical Sciences, Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
| | - Carl De Trez
- Vrije Universiteit Brussel, Laboratory for Cellular and Molecular Immunology (CMIM), Brussels, Belgium
| | - Jean-Pierre Timmermans
- University of Antwerp, Department of Veterinary Sciences, Laboratory of Cell biology & Histology, Wilrijk, Belgium
| | - Sarah Hendrickx
- University of Antwerp, Department of Biomedical Sciences, Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
| | - Louis Maes
- University of Antwerp, Department of Biomedical Sciences, Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
| | - Guy Caljon
- University of Antwerp, Department of Biomedical Sciences, Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
- * E-mail:
| |
Collapse
|
17
|
Filipczak N, Yalamarty SSK, Li X, Parveen F, Torchilin V. Developments in Treatment Methodologies Using Dendrimers for Infectious Diseases. MOLECULES (BASEL, SWITZERLAND) 2021; 26:molecules26113304. [PMID: 34072765 PMCID: PMC8198206 DOI: 10.3390/molecules26113304] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/22/2021] [Accepted: 05/23/2021] [Indexed: 02/02/2023]
Abstract
Dendrimers comprise a specific group of macromolecules, which combine structural properties of both single molecules and long expanded polymers. The three-dimensional form of dendrimers and the extensive possibilities for use of additional substrates for their construction creates a multivalent potential and a wide possibility for medical, diagnostic and environmental purposes. Depending on their composition and structure, dendrimers have been of interest in many fields of science, ranging from chemistry, biotechnology to biochemical applications. These compounds have found wide application from the production of catalysts for their use as antibacterial, antifungal and antiviral agents. Of particular interest are peptide dendrimers as a medium for transport of therapeutic substances: synthetic vaccines against parasites, bacteria and viruses, contrast agents used in MRI, antibodies and genetic material. This review focuses on the description of the current classes of dendrimers, the methodology for their synthesis and briefly drawbacks of their properties and their use as potential therapies against infectious diseases.
Collapse
Affiliation(s)
- Nina Filipczak
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA; (N.F.); (S.S.K.Y.); (X.L.); (F.P.)
| | - Satya Siva Kishan Yalamarty
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA; (N.F.); (S.S.K.Y.); (X.L.); (F.P.)
| | - Xiang Li
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA; (N.F.); (S.S.K.Y.); (X.L.); (F.P.)
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Farzana Parveen
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA; (N.F.); (S.S.K.Y.); (X.L.); (F.P.)
- The Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Vladimir Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA; (N.F.); (S.S.K.Y.); (X.L.); (F.P.)
- Department of Oncology, Radiotherapy and Plastic Surgery, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
- Correspondence:
| |
Collapse
|
18
|
Maksouri H, Darif D, Estaquier J, Riyad M, Desterke C, Lemrani M, Dang PMC, Akarid K. The Modulation of NADPH Oxidase Activity in Human Neutrophils by Moroccan Strains of Leishmania major and Leishmania tropica Is Not Associated with p47 phox Phosphorylation. Microorganisms 2021; 9:1025. [PMID: 34068760 PMCID: PMC8151549 DOI: 10.3390/microorganisms9051025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 11/30/2022] Open
Abstract
Polymorphonuclear neutrophils (PMNs) are the first phagocyte recruited and infected by Leishmania. They synthetize superoxide anions (O2-) under the control of the NADPH oxidase complex. In Morocco, Leishmania major and L. tropica are the main species responsible for cutaneous leishmaniasis (CL). The impact of these parasites on human PMN functions is still unclear. We evaluated the in vitro capacity of primary Moroccan strains of L. major and L. tropica to modulate PMN O2- production and p47phox phosphorylation status of the NADPH oxidase complex. PMNs were isolated from healthy blood donors, and their infection rate was measured by microscopy. O2- production was measured by superoxide dismutase-inhibitable reduction of cytochrome C. P47phox phosphorylation was analyzed by Western blot using specific antibodies against Ser328 and Ser345 sites. Whereas we did not observe any difference in PMN infectivity rate, our results indicated that only L. tropica promastigotes inhibited both fMLF- and PMA-mediated O2- production independently of p47phox phosphorylation. Leishmania soluble antigens (SLAs) from both species significantly inhibited O2- induced by fMLF or PMA. However, they only decreased PMA-induced p47phox phosphorylation. L. major and L. tropica modulated differently O2- production by human PMNs independently of p47phox phosphorylation. The inhibition of ROS production by L. tropica could be a mechanism of its survival within PMNs that might explain the reported chronic pathogenicity of L. tropica CL.
Collapse
Affiliation(s)
- Hasnaa Maksouri
- Research Team on Immunopathology of Infectious and Systemic Diseases, Laboratory of Cellular and Molecular Pathology, Faculty of Medicine and Pharmacy (FMPC), Hassan II University of Casablanca (UH2C), 20000 Casablanca, Morocco; (H.M.); (M.R.)
- Molecular Genetics and Immunophysiopathology Research Team, Health and Environment Laboratory, Aïn Chock Faculty of Sciences, UH2C, 20000 Casablanca, Morocco;
| | - Dounia Darif
- Molecular Genetics and Immunophysiopathology Research Team, Health and Environment Laboratory, Aïn Chock Faculty of Sciences, UH2C, 20000 Casablanca, Morocco;
| | - Jerome Estaquier
- INSERM U1124, Paris University, 75006 Paris, France
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Medicine, Laval University, Quebec City, QC G1V0A6, Canada
| | - Myriam Riyad
- Research Team on Immunopathology of Infectious and Systemic Diseases, Laboratory of Cellular and Molecular Pathology, Faculty of Medicine and Pharmacy (FMPC), Hassan II University of Casablanca (UH2C), 20000 Casablanca, Morocco; (H.M.); (M.R.)
| | - Christophe Desterke
- Faculty of Medicine of the Kremlin-Bicêtre, University Paris-Sud, 94270 Paris, France;
| | - Meryem Lemrani
- Laboratory of Parasitology and Vector-Borne-Diseases, Institut Pasteur du Maroc, 20250 Casablanca, Morocco;
| | - Pham My-Chan Dang
- INSERM-U1149, CNRS-ERL8252, Inflammation Research Center, 75018 Paris, France;
- Inflamex Laboratory of Excellence, Faculty of Medicine, Site Xavier Bichat, University of Paris, 75018 Paris, France
| | - Khadija Akarid
- Molecular Genetics and Immunophysiopathology Research Team, Health and Environment Laboratory, Aïn Chock Faculty of Sciences, UH2C, 20000 Casablanca, Morocco;
| |
Collapse
|
19
|
Elmahallawy EK, Alkhaldi AAM, Saleh AA. Host immune response against leishmaniasis and parasite persistence strategies: A review and assessment of recent research. Biomed Pharmacother 2021; 139:111671. [PMID: 33957562 DOI: 10.1016/j.biopha.2021.111671] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 04/18/2021] [Accepted: 04/24/2021] [Indexed: 12/13/2022] Open
Abstract
Leishmaniasis, a neglected parasitic disease caused by a unicellular protozoan of the genus Leishmania, is transmitted through the bite of a female sandfly. The disease remains a major public health problem and is linked to tropical and subtropical regions, with an endemic picture in several regions, including East Africa, the Mediterranean basin and South America. The different causative species display a diversity of clinical presentations; therefore, the immunological data on leishmaniasis are both scarce and controversial for the different forms and infecting species of the parasite. The present review highlights the main immune parameters associated with leishmaniasis that might contribute to a better understanding of the pathogenicity of the parasite and the clinical outcomes of the disease. Our aim was to provide a concise overview of the immunobiology of the disease and the factors that influence it, as this knowledge may be helpful in developing novel chemotherapeutic and vaccine strategies.
Collapse
Affiliation(s)
- Ehab Kotb Elmahallawy
- Department of Zoonoses, Faculty of Veterinary Medicine, Sohag University, Sohag 82524, Egypt.
| | | | - Amira A Saleh
- Department of Medical Parasitology, Faculty of Medicine, Zagazig University, Zgazig, Egypt
| |
Collapse
|
20
|
Ohms M, Ferreira C, Busch H, Wohlers I, Guerra de Souza AC, Silvestre R, Laskay T. Enhanced Glycolysis Is Required for Antileishmanial Functions of Neutrophils Upon Infection With Leishmania donovani. Front Immunol 2021; 12:632512. [PMID: 33815385 PMCID: PMC8017142 DOI: 10.3389/fimmu.2021.632512] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/02/2021] [Indexed: 01/16/2023] Open
Abstract
Visceral leishmaniasis (VL) is a fatal parasitic disease if untreated. Treatment options of VL diminish due to emerging drug resistance. Although the principal host cells for the multiplication of Leishmania are macrophages, neutrophils are the first cells infected with the parasites rapidly after parasite inoculation. Leishmania can survive in neutrophils despite the potent antimicrobial effector functions of neutrophils that can eliminate the parasites. Recently, the growing field of immunometabolism provided strong evidence for the therapeutic potential in targeting metabolic processes as a means of controlling immune effector functions. Therefore, the understanding of the immunometabolic profile of neutrophils during Leishmania infection could provide new promising targets for host-directed therapies against VL. To our knowledge, this is the first study addressing the bioenergetics profile of L. donovani-infected primary human neutrophils. Transcriptome analysis of L. donovani-infected neutrophils revealed an early significant upregulation of several glycolytic enzymes. Extracellular flux analysis showed that glycolysis and glycolytic capacity were upregulated in L. donovani-infected neutrophils at 6 h post infection. An increased glucose uptake and accumulation of glycolytic end products were further signs for an elevated glycolytic metabolism in L. donovani-infected neutrophils. At the same time point, oxidative phosphorylation provided NADPH for the oxidative burst but did not contribute to ATP production. Inhibition of glycolysis with 2-DG significantly reduced the survival of L. donovani promastigotes in neutrophils and in culture. However, this reduction was due to a direct antileishmanial effect of 2-DG and not a consequence of enhanced antileishmanial activity of neutrophils. To further address the impact of glucose metabolism during the first days of infection in vivo, we treated C57BL/6 mice with 2-DG prior to infection with L. donovani and assessed the parasite load one day and seven days post infection. Our results show, that seven days post-infection the parasite load of 2-DG treated animals was significantly higher than in mock treated animals. This data indicates that glycolysis serves as major energy source for antimicrobial effector functions against L. donovani. Inhibition of glycolysis abrogates important neutrophil effector functions that are necessary the initial control of Leishmania infection.
Collapse
Affiliation(s)
- Mareike Ohms
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Carolina Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Guimarães, Portugal
| | - Hauke Busch
- Medical Systems Biology Division, Lübeck Institute of Experimental Dermatology and Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Inken Wohlers
- Medical Systems Biology Division, Lübeck Institute of Experimental Dermatology and Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Ana Cristina Guerra de Souza
- Department of Cell Technology, Fraunhofer Research Institution for Marine Biotechnology and Cell Technology (EMB), Lübeck, Germany
| | - Ricardo Silvestre
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Guimarães, Portugal
| | - Tamás Laskay
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| |
Collapse
|
21
|
Alcoforado Diniz J, Chaves MM, Vaselek S, Miserani Magalhães RD, Ricci-Azevedo R, de Carvalho RVH, Lorenzon LB, Ferreira TR, Zamboni D, Walrad PB, Volf P, Sacks DL, Cruz AK. Protein methyltransferase 7 deficiency in Leishmania major increases neutrophil associated pathology in murine model. PLoS Negl Trop Dis 2021; 15:e0009230. [PMID: 33651805 PMCID: PMC7954300 DOI: 10.1371/journal.pntd.0009230] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 03/12/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023] Open
Abstract
Leishmania major is the main causative agent of cutaneous leishmaniasis in the Old World. In Leishmania parasites, the lack of transcriptional control is mostly compensated by post-transcriptional mechanisms. Methylation of arginine is a conserved post-translational modification executed by Protein Arginine Methyltransferase (PRMTs). The genome from L. major encodes five PRMT homologs, including the cytosolic protein associated with several RNA-binding proteins, LmjPRMT7. It has been previously reported that LmjPRMT7 could impact parasite infectivity. In addition, a more recent work has clearly shown the importance of LmjPRMT7 in RNA-binding capacity and protein stability of methylation targets, demonstrating the role of this enzyme as an important epigenetic regulator of mRNA metabolism. In this study, we unveil the impact of PRMT7-mediated methylation on parasite development and virulence. Our data reveals that higher levels of LmjPRMT7 can impair parasite pathogenicity, and that deletion of this enzyme rescues the pathogenic phenotype of an attenuated strain of L. major. Interestingly, lesion formation caused by LmjPRMT7 knockout parasites is associated with an exacerbated inflammatory reaction in the tissue correlated with an excessive neutrophil recruitment. Moreover, the absence of LmjPRMT7 also impairs parasite development within the sand fly vector Phlebotomus duboscqi. Finally, a transcriptome analysis shed light onto possible genes affected by depletion of this enzyme. Taken together, this study highlights how post-transcriptional regulation can affect different aspects of the parasite biology. Understanding the genetics of Leishmania, a protozoan parasite causing leishmaniasis, is relevant for understanding fundamental questions on the pathogen’s biology and its interaction with hosts. We explore mechanisms used by Leishmania to promptly adapt to different hosts investigating the control of gene expression occurring at the post-transcriptional level in the parasite. Methylation of arginine performed by Protein Arginine Methyltransferase (PRMTs), among other post-translational modifications, may alter the function and interactions of target proteins, some of them are RNA binding proteins, known regulators of gene expression. In this study, we unveil the impact of PRMT7 on parasite development and pathogenicity. In addition to a negative correlation between the levels of LmjPRMT7 and parasite pathogenicity, we observed an impairment of the parasite development in the sand fly vector. Remarkably, despite a severe lesion development in mice, we observed no differences in parasite burden between infections with the pathogenic LmjPRMT7 knockout parasite or the attenuated parental line. Instead, the severe pathology observed is associated with an exacerbated inflammatory response correlated with excessive neutrophil recruitment.
Collapse
Affiliation(s)
- Juliana Alcoforado Diniz
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Mariana M. Chaves
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Slavica Vaselek
- Department of Parasitology, Charles University, Prague, Czech Republic
| | - Rubens D. Miserani Magalhães
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Rafael Ricci-Azevedo
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Renan V. H. de Carvalho
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Lucas B. Lorenzon
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Tiago R. Ferreira
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Dario Zamboni
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - Petr Volf
- Department of Parasitology, Charles University, Prague, Czech Republic
| | - David L. Sacks
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Angela K. Cruz
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
22
|
Bogdan C. Macrophages as host, effector and immunoregulatory cells in leishmaniasis: Impact of tissue micro-environment and metabolism. Cytokine X 2020; 2:100041. [PMID: 33604563 PMCID: PMC7885870 DOI: 10.1016/j.cytox.2020.100041] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 12/13/2022] Open
Abstract
Leishmania are protozoan parasites that predominantly reside in myeloid cells within their mammalian hosts. Monocytes and macrophages play a central role in the pathogenesis of all forms of leishmaniasis, including cutaneous and visceral leishmaniasis. The present review will highlight the diverse roles of macrophages in leishmaniasis as initial replicative niche, antimicrobial effectors, immunoregulators and as safe hideaway for parasites persisting after clinical cure. These multiplex activities are either ascribed to defined subpopulations of macrophages (e.g., Ly6ChighCCR2+ inflammatory monocytes/monocyte-derived dendritic cells) or result from different activation statuses of tissue macrophages (e.g., macrophages carrying markers of of classical [M1] or alternative activation [M2]). The latter are shaped by immune- and stromal cell-derived cytokines (e.g., IFN-γ, IL-4, IL-10, TGF-β), micro milieu factors (e.g., hypoxia, tonicity, amino acid availability), host cell-derived enzymes, secretory products and metabolites (e.g., heme oxygenase-1, arginase 1, indoleamine 2,3-dioxygenase, NOS2/NO, NOX2/ROS, lipids) as well as by parasite products (e.g., leishmanolysin/gp63, lipophosphoglycan). Exciting avenues of current research address the transcriptional, epigenetic and translational reprogramming of macrophages in a Leishmania species- and tissue context-dependent manner.
Collapse
Key Words
- (L)CL, (localized) cutaneous leishmaniasis
- AHR, aryl hydrocarbon receptor
- AMP, antimicrobial peptide
- Arg, arginase
- Arginase
- CAMP, cathelicidin-type antimicrobial peptide
- CR, complement receptor
- DC, dendritic cells
- DCL, diffuse cutaneous leishmaniasis
- HO-1, heme oxygenase 1
- Hypoxia
- IDO, indoleamine-2,3-dioxygenase
- IFN, interferon
- IFNAR, type I IFN (IFN-α/β) receptor
- IL, interleukin
- Interferon-α/β
- Interferon-γ
- JAK, Janus kinase
- LPG, lipophosphoglycan
- LRV1, Leishmania RNA virus 1
- Leishmaniasis
- Macrophages
- Metabolism
- NCX1, Na+/Ca2+ exchanger 1
- NFAT5, nuclear factor of activated T cells 5
- NK cell, natural killer cell
- NO, nitric oxide
- NOS2 (iNOS), type 2 (or inducible) nitric oxide synthase
- NOX2, NADPH oxidase 2 (gp91 or cytochrome b558 β-subunit of Phox)
- Nitric oxide
- OXPHOS, mitochondrial oxidative phosphorylation
- PKDL, post kala-azar dermal leishmaniasis
- Phagocyte NADPH oxidase
- Phox, phagocyte NADPH oxidase
- RNS, reactive nitrogen species
- ROS, reactive oxygen species
- SOCS, suppressor of cytokine signaling
- STAT, signal transducer and activator of transcription
- TGF-β, transforming growth factor-beta
- TLR, toll-like receptor
- Th1 (Th2), type 1 (type2) T helper cell
- Tonicity
- VL, visceral leishmaniasis
- mTOR, mammalian/mechanistic target of rapamycin
Collapse
Affiliation(s)
- Christian Bogdan
- Mikrobiologisches Institut - klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany.,Medical Immunology Campus Erlangen, FAU Erlangen-Nürnberg, D-91054 Erlangen, Germany
| |
Collapse
|
23
|
Masoudzadeh N, Östensson M, Persson J, Mashayekhi Goyonlo V, Agbajogu C, Taslimi Y, Erfanian Salim R, Zahedifard F, Mizbani A, Malekafzali Ardekani H, Gunn BM, Rafati S, Harandi AM. Molecular signatures of anthroponotic cutaneous leishmaniasis in the lesions of patients infected with Leishmania tropica. Sci Rep 2020; 10:16198. [PMID: 33004861 PMCID: PMC7529897 DOI: 10.1038/s41598-020-72671-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 08/10/2020] [Indexed: 12/23/2022] Open
Abstract
Anthroponotic cutaneous leishmaniasis (CL) caused by Leishmania tropica (L. tropica) represents a public health challenge in several resource poor settings. We herein employed a systems analysis approach to study molecular signatures of CL caused by L. tropica in the skin lesions of ulcerative CL (UCL) and non-ulcerative CL (NUCL) patients. Results from RNA-seq analysis determined shared and unique functional transcriptional pathways in the lesions of the UCL and NUCL patients. Several transcriptional pathways involved in inflammatory response were positively enriched in the CL lesions. A multiplexed inflammatory protein analysis showed differential profiles of inflammatory cytokines and chemokines in the UCL and NUCL lesions. Transcriptional pathways for Fcγ receptor dependent phagocytosis were among shared enriched pathways. Using L. tropica specific antibody (Ab)-mediated phagocytosis assays, we could substantiate Ab-dependent cellular phagocytosis (ADCP) and Ab-dependent neutrophil phagocytosis (ADNP) activities in the lesions of the UCL and NUCL patients, which correlated with L. tropica specific IgG Abs. Interestingly, a negative correlation was observed between parasite load and L. tropica specific IgG/ADCP/ADNP in the skin lesions of CL patients. These results enhance our understanding of human skin response to CL caused by L. tropica.
Collapse
Affiliation(s)
- Nasrin Masoudzadeh
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Malin Östensson
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Josefine Persson
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Christopher Agbajogu
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Yasaman Taslimi
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | | | - Farnaz Zahedifard
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | | | | | - Bronwyn M Gunn
- Ragon Institute of MGH, MIT, and Harvard University, Cambridge, MA, 02139, USA
- Paul G. Allen School of Global Animal Health, Washington State University, Pullman, WA, 99164, USA
| | - Sima Rafati
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran.
| | - Ali M Harandi
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, The University of British Columbia, Vancouver, Canada.
| |
Collapse
|
24
|
Vitamin D increases killing of intracellular Leishmania amazonensis in vitro independently of macrophage oxidative mechanisms. Parasitology 2020; 147:1792-1800. [PMID: 32958098 DOI: 10.1017/s0031182020001791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Vitamin D has been reported to activate macrophage microbicidal mechanisms by inducing the production of antimicrobial peptides and nitric oxide (NO), but conversely has been shown to contribute to a greater susceptibility to Leishmania amazonensis infection in mice. Thus, this study aimed to evaluate the role of vitamin D during intracellular infection with L. amazonensis by examining its effect on macrophage oxidative mechanisms and parasite survival in vitro. Vitamins D2 and D3 significantly inhibited promastigote and amastigote growth in vitro. Vitamin D3 was not able to induce NO and reactive oxygen species (ROS) production in uninfected macrophages or macrophages infected with L. amazonensis. In addition, vitamin D3 in combination with interferon (IFN)-γ did not enhance amastigote killing and in fact, significantly reduced NO and ROS production when compared with the effect of IFN-γ alone. In this study, we demonstrated that vitamin D directly reduces parasite growth in infected macrophages (approximately 50-60% at 50 μm) but this effect is independent of the activation of macrophage oxidative mechanisms. These findings will contribute to a better understanding of the role of vitamin D in cutaneous leishmaniasis.
Collapse
|
25
|
Alfituri OA, Quintana JF, MacLeod A, Garside P, Benson RA, Brewer JM, Mabbott NA, Morrison LJ, Capewell P. To the Skin and Beyond: The Immune Response to African Trypanosomes as They Enter and Exit the Vertebrate Host. Front Immunol 2020; 11:1250. [PMID: 32595652 PMCID: PMC7304505 DOI: 10.3389/fimmu.2020.01250] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/18/2020] [Indexed: 12/14/2022] Open
Abstract
African trypanosomes are single-celled extracellular protozoan parasites transmitted by tsetse fly vectors across sub-Saharan Africa, causing serious disease in both humans and animals. Mammalian infections begin when the tsetse fly penetrates the skin in order to take a blood meal, depositing trypanosomes into the dermal layer. Similarly, onward transmission occurs when differentiated and insect pre-adapted forms are ingested by the fly during a blood meal. Between these transmission steps, trypanosomes access the systemic circulation of the vertebrate host via the skin-draining lymph nodes, disseminating into multiple tissues and organs, and establishing chronic, and long-lasting infections. However, most studies of the immunobiology of African trypanosomes have been conducted under experimental conditions that bypass the skin as a route for systemic dissemination (typically via intraperitoneal or intravenous routes). Therefore, the importance of these initial interactions between trypanosomes and the skin at the site of initial infection, and the implications for these processes in infection establishment, have largely been overlooked. Recent studies have also demonstrated active and complex interactions between the mammalian host and trypanosomes in the skin during initial infection and revealed the skin as an overlooked anatomical reservoir for transmission. This highlights the importance of this organ when investigating the biology of trypanosome infections and the associated immune responses at the initial site of infection. Here, we review the mechanisms involved in establishing African trypanosome infections and potential of the skin as a reservoir, the role of innate immune cells in the skin during initial infection, and the subsequent immune interactions as the parasites migrate from the skin. We suggest that a thorough identification of the mechanisms involved in establishing African trypanosome infections in the skin and their progression through the host is essential for the development of novel approaches to interrupt disease transmission and control these important diseases.
Collapse
Affiliation(s)
- Omar A. Alfituri
- Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Juan F. Quintana
- Wellcome Centre for Integrative Parasitology, College of Medical, Veterinary and Life Sciences, Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Annette MacLeod
- Wellcome Centre for Integrative Parasitology, College of Medical, Veterinary and Life Sciences, Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Paul Garside
- Wellcome Centre for Integrative Parasitology, College of Medical, Veterinary and Life Sciences, Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Robert A. Benson
- Wellcome Centre for Integrative Parasitology, College of Medical, Veterinary and Life Sciences, Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, United Kingdom
| | - James M. Brewer
- Wellcome Centre for Integrative Parasitology, College of Medical, Veterinary and Life Sciences, Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Neil A. Mabbott
- Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Liam J. Morrison
- Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Paul Capewell
- College of Medical, Veterinary and Life Sciences, Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
26
|
Toepp AJ, Petersen CA. The balancing act: Immunology of leishmaniosis. Res Vet Sci 2020; 130:19-25. [PMID: 32109759 DOI: 10.1016/j.rvsc.2020.02.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 01/29/2020] [Accepted: 02/10/2020] [Indexed: 12/12/2022]
Abstract
Immune control of Leishmania infantum, the causative agent of most canine leishmaniosis (CanL), requires a balancing act between inflammatory and regulatory responses. This balance is specifically between the proinflammatory T helper 1 type (Th1) CD4+ T cells that are responsible for controlling parasite replication and T regulatory 1 cells which mediate an immunosuppressive, regulatory, response needed to dampen overabundant inflammation but if predominant, result in CanL progression. How this delicate immune cell interaction occurs in the dog will be highlighted in this review, focusing on the progressive changes observed within myeloid lineage cells (predominantly macrophages), B cells and T cells. After exposure to parasites, macrophages should become activated, eliminating L. infantum through release of reactive oxygen species. Unfortunately, multiple parasite and host factors can prevent macrophage activation allowing parasites to persist within them. T cells balance between a productive TH1 type CD4+ response capable of producing IFN-γ which aids macrophage activation versus T cell exhaustion which reduces T cell proliferation, IFN-γ production and allows parasite expansion within macrophages. Neutrophils and Th17 cells add to the inflammatory state, aiding in parasite removal, but also leading to pathology. A regulatory B cell population increases IL-10 production and down regulates the TH1 response allowing parasite growth. All of these immune challenges affect the balance between progression to clinical disease and maintaining sub-clinical disease. Vaccines and immunotherapies targeted at recovering or maintaining T and B cell function can be important factors in mending the immune balance required to survive CanL.
Collapse
Affiliation(s)
- Angela J Toepp
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA; Center for Emerging Infectious Diseases, University of Iowa Research Park, Coralville, IA 52241, USA
| | - Christine A Petersen
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA; Center for Emerging Infectious Diseases, University of Iowa Research Park, Coralville, IA 52241, USA.
| |
Collapse
|
27
|
Tissue-resident macrophages can be generated de novo in adult human skin from resident progenitor cells during substance P-mediated neurogenic inflammation ex vivo. PLoS One 2020; 15:e0227817. [PMID: 31971954 PMCID: PMC6977738 DOI: 10.1371/journal.pone.0227817] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 12/30/2019] [Indexed: 12/11/2022] Open
Abstract
Besides monocyte (MO)-derived macrophages (MACs), self-renewing tissue-resident macrophages (trMACs) maintain the intracutaneous MAC pool in murine skin. Here, we have asked whether the same phenomenon occurs in human skin using organ-cultured, full-thickness skin detached from blood circulation and bone marrow. Skin stimulation ex vivo with the neuropeptide substance P (SP), mimicking neurogenic skin inflammation, significantly increased the number of CD68+MACs in the papillary dermis without altering intracutaneous MAC proliferation or apoptosis. Since intraluminal CD14+MOs were undetectable in the non-perfused dermal vasculature, new MACs must have differentiated from resident intracutaneous progenitor cells in human skin. Interestingly, CD68+MACs were often seen in direct cell-cell-contact with cells expressing both, the hematopoietic stem cell marker CD34 and SP receptor (neurokinin-1 receptor [NK1R]). These cell-cell contacts and CD34+cell proliferation were up-regulated in SP-treated skin samples. Collectively, our study provides the first evidence that resident MAC progenitors, from which mature MACs can rapidly differentiate within the tissue, do exist in normal adult human skin. That these NK1R+trMAC-progenitor cells quickly respond to a key stress-associated neuroinflammatory stimulus suggests that this may satisfy increased local MAC demand under conditions of wounding/stress.
Collapse
|
28
|
Cutaneous Leishmaniasis: The Complexity of Host's Effective Immune Response against a Polymorphic Parasitic Disease. J Immunol Res 2019; 2019:2603730. [PMID: 31871953 PMCID: PMC6913332 DOI: 10.1155/2019/2603730] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 10/05/2019] [Accepted: 10/22/2019] [Indexed: 12/14/2022] Open
Abstract
This review is aimed at providing a comprehensive outline of the immune response displayed against cutaneous leishmaniasis (CL), the more common zoonotic infection caused by protozoan parasites of the genus Leishmania. Although of polymorphic clinical presentation, classically CL is characterized by leishmaniotic lesions on the face and extremities of the patients, which can be ulcerative, and even after healing can lead to permanent injuries and disfigurement, affecting significantly their psychological, social, and economic well-being. According a report released by the World Health Organization, the disability-adjusted life years (DALYs) lost due to leishmaniasis are close to 2.4 million, annually there are 1.0–1.5 million new cases of CL, and a numerous population is at risk in the endemic areas. Despite its increasing worldwide incidence, it is one of the so-called neglected tropical diseases. Furthermore, this review provides an overview of the existing knowledge of the host innate and acquired immune response to cutaneous species of Leishmania. The use of animal models and of in vitro studies has improved the understanding of parasite-host interplay and the complexity of immune mechanisms involved. The importance of diagnosis accuracy associated with effective patient management in CL reduction is highlighted. However, the multiple factors involved in CL epizoology associated with the unavailability of vaccines or drugs to prevent infection make difficult to formulate an effective strategy for CL control.
Collapse
|
29
|
Wei F, Gong W, Wang J, Yang Y, Liu J, Wang Y, Cao J. Role of the lipoxin A4 receptor in the development of neutrophil extracellular traps in Leishmania infantum infection. Parasit Vectors 2019; 12:275. [PMID: 31142352 PMCID: PMC6542009 DOI: 10.1186/s13071-019-3530-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 05/23/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Neutrophils play an immunomodulatory role through the release of neutrophil extracellular traps (NETs). NETs are released in response to Leishmania infection, but the mechanism of NET extrusion has not been elucidated. The lipoxin A4 receptor on neutrophils is crucial for the inflammatory response and immune regulation of many diseases, including Leishmania infection. Therefore, in the present study, we tried to explore whether Leishmania infantum promastigotes stimulate neutrophil activation and NET release via activating the lipoxin A4 receptor. RESULTS Leishmania infantum promastigotes stimulated neutrophil activity, but blocking of the lipoxin A4 receptor with its antagonist Boc prior to L. infantum stimulation abrogated these effects. Neutrophils showed citrullinated histone H3 expression and simultaneous NET extrusion on L. infantum stimulation, but a decline in both was observed on blocking of the lipoxin A4 receptor. Moreover, differentiated HL-60 cells with lipoxin A4 receptor silencing showed a decrease in citrullinated histone H3 expression as compared to the unsilenced HL-60 samples on stimulation with promastigotes. CONCLUSIONS Leishmania infantum promastigotes altered the characteristics of neutrophils and induced NET extrusion by activating the lipoxin A4 receptor. The lipoxin A4 receptor may have potential as a therapeutic target in relation to NET extrusion in the treatment of leishmaniasis, but its mechanisms of action need to be explored in more depth.
Collapse
Affiliation(s)
- Furong Wei
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, China.,Chinese Center for Tropical Diseases Research, Shanghai, 200025, China.,WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, 200025, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025, China
| | - Wenci Gong
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, China.,Chinese Center for Tropical Diseases Research, Shanghai, 200025, China.,WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, 200025, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025, China
| | - Junyun Wang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, China.,Chinese Center for Tropical Diseases Research, Shanghai, 200025, China.,WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, 200025, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025, China
| | - Yuetao Yang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, China.,Chinese Center for Tropical Diseases Research, Shanghai, 200025, China.,WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, 200025, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025, China
| | - Jianxiu Liu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, China.,Chinese Center for Tropical Diseases Research, Shanghai, 200025, China.,WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, 200025, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025, China
| | - Yanjuan Wang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, China.,Chinese Center for Tropical Diseases Research, Shanghai, 200025, China.,WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, 200025, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025, China
| | - Jianping Cao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, China. .,Chinese Center for Tropical Diseases Research, Shanghai, 200025, China. .,WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, China. .,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, 200025, China. .,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025, China.
| |
Collapse
|
30
|
Oualha R, Barhoumi M, Marzouki S, Harigua-Souiai E, Ben Ahmed M, Guizani I. Infection of Human Neutrophils With Leishmania infantum or Leishmania major Strains Triggers Activation and Differential Cytokines Release. Front Cell Infect Microbiol 2019; 9:153. [PMID: 31134162 PMCID: PMC6524560 DOI: 10.3389/fcimb.2019.00153] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/24/2019] [Indexed: 12/27/2022] Open
Abstract
Leishmaniases are neglected diseases, caused by intracellular protozoan parasites of the Leishmania (L.) genus. Although the principal host cells of the parasites are macrophages, neutrophils are the first cells rapidly recruited to the site of parasites inoculation, where they play an important role in the early recognition and elimination of the parasites. The nature of early interactions between neutrophils and Leishmania could influence the outcome of infection. Herein we aimed to evaluate whether different Leishmania strains, responsible for distinct clinical manifestations, could influence ex vivo functional activity of neutrophils. Human polymorphonuclear leukocytes were isolated from 14 healthy volunteers and the ex vivo infection of these cells was done with two L. infantum and one L. major strains. Infection parameters were determined and neutrophils activation was assessed by oxidative burst, degranulation, DNA release and apoptosis; cytokine production was measured by a multiplex flow cytometry analysis. Intracellular amastigotes were rescued to determine Leishmania strains survival. The results showed that L. infantum and L. major promastigotes similarly infected the neutrophils. Oxidative burst, neutrophil elastase, myeloperoxidase activity and apoptosis were significantly increased in infected neutrophils but with no differences between strains. The L. infantum-infected neutrophils induced more DNA release than those infected by L. major. Furthermore, Leishmania strains induced high amounts of IL-8 and stimulated the production of IL-1β, TNF-α, and TGF-β by human neutrophils. We observed that only one strain promoted IL-6 release by these neutrophils. The production of TNF-α was also differently induced by the parasites strains. All these results demonstrate that L. infantum and L. major strains were able to induce globally a similar ex vivo activation and apoptosis of neutrophils; however, they differentially triggered cytokines release from these cells. In addition, rescue of intracellular parasites indicated different survival rates further emphasizing on the influence of parasite strains within a species on the fate of infection.
Collapse
Affiliation(s)
- Rafeh Oualha
- Laboratory of Molecular Epidemiology and Experimental Pathology - LR16IPT04, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia.,Faculté des Sciences de Bizerte, Université de Carthage, Tunis, Tunisia
| | - Mourad Barhoumi
- Laboratory of Molecular Epidemiology and Experimental Pathology - LR16IPT04, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Soumaya Marzouki
- Laboratory of Transmission, Control and Immunobiology of Infections - LR16IPT02, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Emna Harigua-Souiai
- Laboratory of Molecular Epidemiology and Experimental Pathology - LR16IPT04, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Melika Ben Ahmed
- Laboratory of Transmission, Control and Immunobiology of Infections - LR16IPT02, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Ikram Guizani
- Laboratory of Molecular Epidemiology and Experimental Pathology - LR16IPT04, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
31
|
Pitale DM, Gendalur NS, Descoteaux A, Shaha C. Leishmania donovani Induces Autophagy in Human Blood–Derived Neutrophils. THE JOURNAL OF IMMUNOLOGY 2019; 202:1163-1175. [DOI: 10.4049/jimmunol.1801053] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 12/10/2018] [Indexed: 12/19/2022]
|
32
|
Ronet C, Passelli K, Charmoy M, Scarpellino L, Myburgh E, Hauyon La Torre Y, Turco S, Mottram JC, Fasel N, Luther SA, Beverley SM, Launois P, Tacchini-Cottier F. TLR2 Signaling in Skin Nonhematopoietic Cells Induces Early Neutrophil Recruitment in Response to Leishmania major Infection. J Invest Dermatol 2018; 139:1318-1328. [PMID: 30594488 DOI: 10.1016/j.jid.2018.12.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 11/14/2018] [Accepted: 12/05/2018] [Indexed: 12/14/2022]
Abstract
Neutrophils are rapidly recruited to the mammalian skin in response to infection with the cutaneous Leishmania pathogen. The parasites use neutrophils to establish the disease; however, the signals driving early neutrophil recruitment are poorly known. Here, we identified the functional importance of TLR2 signaling in this process. Using bone marrow chimeras and immunohistology, we identified the TLR2-expressing cells involved in this early neutrophil recruitment to be of nonhematopoietic origin. Keratinocytes are damaged and briefly in contact with the parasites during infection. We show that TLR2 triggering by Leishmania major is required for their secretion of neutrophil-attracting chemokines. Furthermore, TLR2 triggering by L. major phosphoglycans is critical for neutrophil recruitment to negatively affect disease development, as shown by better control of lesion size and parasite load in Tlr2-/- compared with wild-type infected mice. Conversely, restoring early neutrophil presence in Tlr2-/- mice through injection of wild-type neutrophils or CXCL1 at the onset of infection resulted in delayed disease resolution comparable to that observed in wild-type mice. Taken together, our data show a crucial role for TLR2-expressing nonhematopoietic skin cells in the recruitment of the first wave of neutrophils after L. major infection, a process that delays disease control.
Collapse
Affiliation(s)
- Catherine Ronet
- Department of Biochemistry, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland; World Health Organization Immunology Research and Training Center, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland
| | - Katiuska Passelli
- Department of Biochemistry, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland; World Health Organization Immunology Research and Training Center, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland
| | - Mélanie Charmoy
- Department of Biochemistry, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland; World Health Organization Immunology Research and Training Center, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland
| | - Leo Scarpellino
- Department of Biochemistry, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland
| | - Elmarie Myburgh
- Centre for Immunology and Infection, Department of Biology, University of York, Heslington, York, UK
| | - Yazmin Hauyon La Torre
- Department of Biochemistry, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland; World Health Organization Immunology Research and Training Center, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland
| | - Salvatore Turco
- Department of Biochemistry, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Jeremy C Mottram
- Centre for Immunology and Infection, Department of Biology, University of York, Heslington, York, UK
| | - Nicolas Fasel
- Department of Biochemistry, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland
| | - Sanjiv A Luther
- Department of Biochemistry, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland
| | - Stephen M Beverley
- Molecular Microbiology Department, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Pascal Launois
- Department of Biochemistry, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland; World Health Organization Immunology Research and Training Center, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland
| | - Fabienne Tacchini-Cottier
- Department of Biochemistry, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland; World Health Organization Immunology Research and Training Center, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland.
| |
Collapse
|
33
|
Muxel SM, Acuña SM, Aoki JI, Zampieri RA, Floeter-Winter LM. Toll-Like Receptor and miRNA-let-7e Expression Alter the Inflammatory Response in Leishmania amazonensis-Infected Macrophages. Front Immunol 2018; 9:2792. [PMID: 30555476 PMCID: PMC6283264 DOI: 10.3389/fimmu.2018.02792] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 11/13/2018] [Indexed: 12/12/2022] Open
Abstract
Parasite recognition by Toll-like receptors (TLRs) contributes to macrophage activation and subsequent control of Leishmania infection through the coordinated production of pro-inflammatory and microbicidal effector molecules. The modulation of microRNA (miRNA) expression by Leishmania infection potentially mediates the post-transcriptional regulation of the expression of genes involved in leishmanicidal activity. Here, the contribution of TLR signaling to the miRNA profile and gene expression was evaluated in Leishmania amazonensis-infected murine macrophages. The infectivity of L. amazonensis was higher in murine bone marrow-derived macrophages from mice knockout for myeloid differentiation factor 88 (MyD88−/−), TLR2 (TLR2−/−), or TLR4 (TLR4−/−) than wild type C57BL/6 (WT). L. amazonensis infection of WT macrophages modulated the expression of 32% of the miRNAs analyzed, while 50% were upregulated. The absence of MyD88, TLR2, and TLR4 altered the percentage of miRNAs modulated during L. amazonensis infection, including the downregulation of let-7e expression. Moreover, the absence of signals mediated by MyD88, TLR2, or TLR4 reduced nitric oxide synthase 2 (Nos2) mRNA expression during infection. Indeed, the inhibition of let-7e increased levels of the Nos2 mRNA and NOS2 (or iNOS) protein and nitric oxide (NO) production in L. amazonensis-infected macrophages (4–24 h). The absence of TLR2 and inhibition of let-7e increased the expression of the arginase 1 (Arg1) mRNA but did not alter the protein level during infection. However, higher levels of the L-arginine transporters Cat2B and Cat1 were detected in the absence of Myd88 signaling during infection but were not altered following let-7e inhibition. The inhibition of let-7e impacted the global expression of genes in the TLR pathway by upregulating the expression of recognition and adaptors molecules, such as Tlr6, Tlr9, Ly96, Tirap, Traf 6, Ticam1, Tollip, Casp8, Map3k1, Mapk8, Nfkbib, Nfkbil1, Ppara, Mapk8ip3, Hspd1, and Ube2n, as well as immunomodulators, such as Ptgs2/Cox2, Csf 2, Csf 3, Ifnb1, Il6ra, and Ilr1, impacting NOS2 expression, NO production and parasite infectiveness. In conclusion, L. amazonensis infection alters the TLR signaling pathways by modulating the expression of miRNAs in macrophages to subvert the host immune responses.
Collapse
Affiliation(s)
- Sandra Marcia Muxel
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Stephanie Maia Acuña
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Juliana Ide Aoki
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Ricardo Andrade Zampieri
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | | |
Collapse
|
34
|
Mhlwatika Z, Aderibigbe BA. Application of Dendrimers for the Treatment of Infectious Diseases. Molecules 2018; 23:E2205. [PMID: 30200314 PMCID: PMC6225509 DOI: 10.3390/molecules23092205] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/14/2018] [Accepted: 06/15/2018] [Indexed: 01/14/2023] Open
Abstract
Dendrimers are drug delivery systems that are characterized by a three-dimensional, star-shaped, branched macromolecular network. They possess ideal properties such as low polydispersity index, biocompatibility and good water solubility. They are made up of the interior and the exterior layers. The exterior layer consists of functional groups that are useful for conjugation of drugs and targeting moieties. The interior layer exhibits improved drug encapsulation efficiency, reduced drug toxicity, and controlled release mechanisms. These unique properties make them useful for drug delivery. Dendrimers have attracted considerable attention as drug delivery system for the treatment of infectious diseases. The treatment of infectious diseases is hampered severely by drug resistance. Several properties of dendrimers such as their ability to overcome drug resistance, toxicity and control the release mechanism of the encapsulated drugs make them ideal systems for the treatment of infectious disease. The aim of this review is to discuss the potentials of dendrimers for the treatment of viral and parasitic infections.
Collapse
Affiliation(s)
- Zandile Mhlwatika
- Department of Chemistry, University of Fort Hare, Alice Campus, Eastern Cape 5700, South Africa.
| | - Blessing Atim Aderibigbe
- Department of Chemistry, University of Fort Hare, Alice Campus, Eastern Cape 5700, South Africa.
| |
Collapse
|
35
|
The Role of Histopathology and Immunohistochemistry in the Diagnosis of Cutaneous Leishmaniasis Without "Discernible" Leishman-Donovan Bodies. Am J Dermatopathol 2018; 39:890-895. [PMID: 29190217 DOI: 10.1097/dad.0000000000000861] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Histopathology plays an important role in the diagnosis of cutaneous leishmaniasis (CL) but Leishman-Donovan (LD) bodies may not always be discernible. Recently, anti-CD1a antibody (Ab), clone MTB1, was found to decorate LD bodies immunohistochemically. OBJECTIVE Can histopathology without discernible LD bodies be used to diagnose CL, and can immunohistochemistry using anti-CD1a Ab, clone MTB1, detect LD bodies in these cases. METHODS Suspected CL lesions were studied histopathologically and immunohistochemically, and the patients' clinical files were reviewed. RESULTS Of the 196 patients with suspected CL, direct smear demonstrated LD bodies in 50 (25.5%). Of the remaining 146 patients, 118 underwent biopsy. In 56 (47.5%) patients, the hematoxylin-eosin-stained sections revealed LD bodies. In 47 (39.8%) patients, LD bodies were not discerned but the histopathology demonstrated histiocytic infiltrates with varying numbers of plasma cells along with other inflammatory cells, and negative Ziehl-Neelsen and periodic acid-Schiff stains. This pattern was termed "histopathology consistent with leishmaniasis." The history, clinical findings, and response to anti-leishmania therapy supported the diagnosis of CL in all of them, and immunostains for CD1a, clone MTB1, detected LD bodies in 11 (23.4%) of these 47 patients. CONCLUSIONS "Histopathology consistent with CL" along with appropriate clinical findings supports the diagnosis of CL in an endemic area, and immunostains with CD1a Ab, clone MTB1, may help in the minority of the cases.
Collapse
|
36
|
Martínez-López M, Soto M, Iborra S, Sancho D. Leishmania Hijacks Myeloid Cells for Immune Escape. Front Microbiol 2018; 9:883. [PMID: 29867798 PMCID: PMC5949370 DOI: 10.3389/fmicb.2018.00883] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 04/17/2018] [Indexed: 12/23/2022] Open
Abstract
Protozoan parasites of the Leishmania genus are the causative agents of leishmaniasis, a group of neglected tropical diseases whose clinical manifestations vary depending on the infectious Leishmania species but also on host factors. Recognition of the parasite by host myeloid immune cells is a key to trigger an effective Leishmania-specific immunity. However, the parasite is able to persist in host myeloid cells by evading, delaying and manipulating host immunity in order to escape host resistance and ensure its transmission. Neutrophils are first in infiltrating infection sites and could act either favoring or protecting against infection, depending on factors such as the genetic background of the host or the parasite species. Macrophages are the main host cells where the parasites grow and divide. However, macrophages are also the main effector population involved in parasite clearance. Parasite elimination by macrophages requires the priming and development of an effector Th1 adaptive immunity driven by specific subtypes of dendritic cells. Herein, we will provide a comprehensive outline of how myeloid cells regulate innate and adaptive immunity against Leishmania, and the mechanisms used by the parasites to promote their evasion and sabotage. Understanding the interactions between Leishmania and the host myeloid cells may lead to the development of new therapeutic approaches and improved vaccination to leishmaniases, an important worldwide health problem in which current therapeutic or preventive approaches are limited.
Collapse
Affiliation(s)
- María Martínez-López
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares "Carlos III", Madrid, Spain
| | - Manuel Soto
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - Salvador Iborra
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares "Carlos III", Madrid, Spain.,Department of Immunology, Complutense University School of Medicine and 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - David Sancho
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares "Carlos III", Madrid, Spain
| |
Collapse
|
37
|
Soulat D, Bogdan C. Function of Macrophage and Parasite Phosphatases in Leishmaniasis. Front Immunol 2017; 8:1838. [PMID: 29312331 PMCID: PMC5743797 DOI: 10.3389/fimmu.2017.01838] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 12/05/2017] [Indexed: 01/23/2023] Open
Abstract
The kinetoplastid protozoan parasites belonging to the genus Leishmania are the causative agents of different clinical forms of leishmaniasis, a vector-borne infectious disease with worldwide prevalence. The protective host immune response against Leishmania parasites relies on myeloid cells such as dendritic cells and macrophages in which upon stimulation by cytokines (e.g., interferon-γ) a complex network of signaling pathways is switched on leading to strong antimicrobial activities directed against the intracellular parasite stage. The regulation of these pathways classically depends on post-translational modifications of proteins, with phosphorylation events playing a cardinal role. Leishmania parasites deactivate their phagocytic host cells by inducing specific mammalian phosphatases that are capable to impede signaling. On the other hand, there is now also evidence that Leishmania spp. themselves express phosphatases that might target host cell molecules and thereby facilitate the intracellular survival of the parasite. This review will present an overview on the modulation of host phosphatases by Leishmania parasites as well as on the known families of Leishmania phosphatases and their possible function as virulence factors. A more detailed understanding of the role of phosphatases in Leishmania–host cell interactions might open new avenues for the treatment of non-healing, progressive forms of leishmaniasis.
Collapse
Affiliation(s)
- Didier Soulat
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany.,Medical Immunology Campus Erlangen, Interdisciplinary Center of the FAU, Erlangen, Germany
| | - Christian Bogdan
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany.,Medical Immunology Campus Erlangen, Interdisciplinary Center of the FAU, Erlangen, Germany
| |
Collapse
|
38
|
Regli IB, Passelli K, Hurrell BP, Tacchini-Cottier F. Survival Mechanisms Used by Some Leishmania Species to Escape Neutrophil Killing. Front Immunol 2017; 8:1558. [PMID: 29250059 PMCID: PMC5715327 DOI: 10.3389/fimmu.2017.01558] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 10/31/2017] [Indexed: 12/27/2022] Open
Abstract
Neutrophils are the most abundant leukocytes in human blood. Upon microbial infection, they are massively and rapidly recruited from the circulation to sites of infection where they efficiently kill pathogens. To this end, neutrophils possess a variety of weapons that can be mobilized and become effective within hours following infection. However, several microbes including some Leishmania spp. have evolved a variety of mechanisms to escape neutrophil killing using these cells as a basis to better invade the host. In addition, neutrophils are also present in unhealing cutaneous lesions where their role remains to be defined. Here, we will review recent progress in the field and discuss the different strategies applied by some Leishmania parasites to escape from being killed by neutrophils and as recently described for Leishmania mexicana, even replicate within these cells. Subversion of neutrophil killing functions by Leishmania is a strategy that allows parasite spreading in the host with a consequent deleterious impact, transforming the primary protective role of neutrophils into a deleterious one.
Collapse
Affiliation(s)
- Ivo B Regli
- Department of Biochemistry, WHO Immunology Research and Training Collaborative Center, University of Lausanne, Lausanne, Switzerland
| | - Katiuska Passelli
- Department of Biochemistry, WHO Immunology Research and Training Collaborative Center, University of Lausanne, Lausanne, Switzerland
| | - Benjamin P Hurrell
- Department of Biochemistry, WHO Immunology Research and Training Collaborative Center, University of Lausanne, Lausanne, Switzerland
| | - Fabienne Tacchini-Cottier
- Department of Biochemistry, WHO Immunology Research and Training Collaborative Center, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
39
|
Gonçalves-de-Albuquerque SDC, Pessoa-e-Silva R, Trajano-Silva LAM, de Goes TC, de Morais RCS, da C. Oliveira CN, de Lorena VMB, de Paiva-Cavalcanti M. The Equivocal Role of Th17 Cells and Neutrophils on Immunopathogenesis of Leishmaniasis. Front Immunol 2017; 8:1437. [PMID: 29163510 PMCID: PMC5670345 DOI: 10.3389/fimmu.2017.01437] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 10/16/2017] [Indexed: 01/17/2023] Open
Abstract
Advances in the understanding of leishmaniasis progression indicate that cellular interactions more complex than the Th1/Th2 paradigm define the course of infection. Th17 cells are a crucial modulator of adaptive immunity against Leishmania parasites acting mainly on neutrophil recruitment and playing a dual role at the site of infection. This review describes the roles of both these cell types in linking innate defense responses to the establishment of specific immunity. We focus on the Th17-neutrophil interaction as a crucial component of anti-Leishmania immunity, and the clinical evolution of cutaneous or visceral leishmaniasis. To date, information obtained through experimental models and patient evaluations suggests that the influence of the presence of interleukin (IL)-17 (the main cytokine produced by Th17 cells) and neutrophils during Leishmania infections is strictly dependent on the tissue (skin or liver/spleen) and parasite species. Also, the time at which neutrophils are recruited, and the persistence of IL-17 in the infection microenvironment, may also be significant. A clearer understanding of these interactions will enable better measurement of the influence of IL-17 and its regulators, and contribute to the identification of disease/resistance biomarkers.
Collapse
Affiliation(s)
| | - Rômulo Pessoa-e-Silva
- Department of Microbiology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | - Lays A. M. Trajano-Silva
- Department of Microbiology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | - Tayná Correia de Goes
- Department of Microbiology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | - Rayana C. S. de Morais
- Department of Microbiology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | - Cíntia N. da C. Oliveira
- Department of Microbiology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | - Virgínia M. B. de Lorena
- Department of Microbiology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | - Milena de Paiva-Cavalcanti
- Department of Microbiology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| |
Collapse
|
40
|
Canine neutrophils activate effector mechanisms in response to Leishmania infantum. Vet Parasitol 2017; 248:10-20. [PMID: 29173534 DOI: 10.1016/j.vetpar.2017.10.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 09/25/2017] [Accepted: 10/17/2017] [Indexed: 12/21/2022]
Abstract
Canine leishmaniosis caused by L. infantum is a severe zoonotic disease. Although macrophages are the definitive host cells, neutrophils are the first cells to encounter the parasite soon after its inoculation in the dermis by the phlebotomine vector. To study the interaction of dog neutrophils and L. infantum promastigotes, blood neutrophils were isolated from healthy donors and the infection was established in vitro. In the majority of the dogs, L. infantum was efficiently phagocytized by neutrophils, and oxidative (superoxide production) and non-oxidative (neutrophil elastase exocytosis) intracellular effector mechanisms were activated, but the release of neutrophil extracellular traps was minimized. Furthermore, promastigotes and culture supernatants induced neutrophil migration, but the prior contact with Leishmania inhibits chemotaxis, which might contribute to neutrophil retention at the inoculation site. Neutrophil-parasite interaction resulted in a decrease in parasite viability, although some intracellular promastigotes survive and maintain their proliferative capacity. These findings indicate that dog neutrophils are competent effector cells able to control the initial L. infantum infection. However, some parasites evade intracellular effector mechanisms and can be transferred to the definitive host cell, the macrophage, contributing to the development of canine leishmaniosis.
Collapse
|
41
|
Sacramento LA, da Costa JL, de Lima MHF, Sampaio PA, Almeida RP, Cunha FQ, Silva JS, Carregaro V. Toll-Like Receptor 2 Is Required for Inflammatory Process Development during Leishmania infantum Infection. Front Microbiol 2017; 8:262. [PMID: 28280488 PMCID: PMC5322192 DOI: 10.3389/fmicb.2017.00262] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 02/07/2017] [Indexed: 12/20/2022] Open
Abstract
Visceral leishmaniasis (VL) is a chronic and fatal disease caused by Leishmania infantum in Brazil. Leukocyte recruitment to infected tissue is a crucial event for the control of infections such as VL. Among inflammatory cells, neutrophils are recruited to the site of Leishmania infection, and these cells may control parasite replication through oxidative or non-oxidative mechanisms. The recruitment, activation and functions of the neutrophils are coordinated by pro-inflammatory cytokines and chemokines during recognition of the parasite by pattern recognition receptors (PRRs). Here, we demonstrated that the Toll-like receptor 2 (TLR2) signaling pathway contributes to the development of the innate immune response during L. infantum infection. The protective mechanism is related to the appropriate recruitment of neutrophils to the inflammatory site. Neutrophil migration is coordinated by DCs that produce CXCL1 and provide a prototypal Th1 and Th17 environment when activated via TLR2. Furthermore, infected TLR2−/− mice failed to induce nitric oxide synthase (iNOS) expression in neutrophils but not in macrophages. In vitro, infected TLR2−/− neutrophils presented deficient iNOS expression, nitric oxide (NO) and TNF-α production, decreased expression of CD11b and reduced L. infantum uptake capacity. The non-responsive state of neutrophils is associated with increased amounts of IL-10. Taken together, these data clarify new mechanisms by which TLR2 functions in promoting the development of the adaptive immune response and effector mechanisms of neutrophils during L. infantum infection.
Collapse
Affiliation(s)
- Laís A Sacramento
- Department of Biochemistry and Immunology, University of São Paulo Ribeirão Preto, Brazil
| | - Jéssica L da Costa
- Department of Biochemistry and Immunology, University of São Paulo Ribeirão Preto, Brazil
| | - Mikhael H F de Lima
- Department of Biochemistry and Immunology, University of São Paulo Ribeirão Preto, Brazil
| | - Pedro A Sampaio
- Department of Biochemistry and Immunology, University of São Paulo Ribeirão Preto, Brazil
| | - Roque P Almeida
- Center for Biology and Health Sciences, Federal University of Sergipe Aracaju, Brazil
| | - Fernando Q Cunha
- Department of Biochemistry and Immunology, University of São PauloRibeirão Preto, Brazil; Department of Pharmacology, Ribeirão Preto Medical School, University of São PauloRibeirão Preto, Brazil
| | - João S Silva
- Department of Biochemistry and Immunology, University of São Paulo Ribeirão Preto, Brazil
| | - Vanessa Carregaro
- Department of Biochemistry and Immunology, University of São Paulo Ribeirão Preto, Brazil
| |
Collapse
|
42
|
Yizengaw E, Getahun M, Tajebe F, Cruz Cervera E, Adem E, Mesfin G, Hailu A, Van der Auwera G, Yardley V, Lemma M, Skhedy Z, Diro E, Yeshanew A, Melkamu R, Mengesha B, Modolell M, Munder M, Müller I, Takele Y, Kropf P. Visceral Leishmaniasis Patients Display Altered Composition and Maturity of Neutrophils as well as Impaired Neutrophil Effector Functions. Front Immunol 2016; 7:517. [PMID: 27965662 PMCID: PMC5126105 DOI: 10.3389/fimmu.2016.00517] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 11/07/2016] [Indexed: 11/13/2022] Open
Abstract
Immunologically, active visceral leishmaniasis (VL) is characterized by profound immunosuppression, severe systemic inflammatory responses, and an impaired capacity to control parasite replication. Neutrophils are highly versatile cells, which play a crucial role in the induction as well as the resolution of inflammation, the control of pathogen replication, and the regulation of immune responses. Neutrophil functions have been investigated in human cutaneous leishmaniasis; however, their role in human VL is poorly understood. In the present study we evaluated the activation status and effector functions of neutrophils in patients with active VL and after successful anti-leishmanial treatment. Our results show that neutrophils are highly activated and have degranulated; high levels of arginase, myeloperoxidase, and elastase, all contained in neutrophils' granules, were found in the plasma of VL patients. In addition, we show that a large proportion of these cells are immature. We also analyzed effector functions of neutrophils that are essential for pathogen clearance and show that neutrophils have an impaired capacity to release neutrophil extracellular traps, produce reactive oxygen species, and phagocytose bacterial particles, but not Leishmania parasites. Our results suggest that impaired effector functions, increased activation, and immaturity of neutrophils play a key role in the pathogenesis of VL.
Collapse
Affiliation(s)
- Endalew Yizengaw
- Department of Immunology, University of Gondar , Gondar , Ethiopia
| | - Mulusew Getahun
- Department of Immunology, University of Gondar , Gondar , Ethiopia
| | | | | | - Emebet Adem
- Leishmaniasis Research and Treatment Centre, Gondar University , Gondar , Ethiopia
| | - Getnet Mesfin
- Department of Immunology, University of Gondar , Gondar , Ethiopia
| | - Asrat Hailu
- Department of Microbiology, Immunology and Parasitology, Addis Ababa University , Addis Ababa , Ethiopia
| | - Gert Van der Auwera
- Department of Biomedical Sciences, Institute of Tropical Medicine , Antwerp , Belgium
| | - Vanessa Yardley
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine , London , UK
| | - Mulualem Lemma
- Department of Internal Medicine, University of Gondar , Gondar , Ethiopia
| | - Ziv Skhedy
- Department of Mathematics and Statistics, University of Hasselt , Hasselt , Belgium
| | - Ermias Diro
- Leishmaniasis Research and Treatment Centre, Gondar University , Gondar , Ethiopia
| | - Arega Yeshanew
- Leishmaniasis Research and Treatment Centre, Gondar University , Gondar , Ethiopia
| | - Roma Melkamu
- Leishmaniasis Research and Treatment Centre, Gondar University , Gondar , Ethiopia
| | - Bewketu Mengesha
- Leishmaniasis Research and Treatment Centre, Gondar University , Gondar , Ethiopia
| | - Manuel Modolell
- Department of Cellular Immunology, Max-Planck-Institute for Immunobiology and Epigenetics , Freiburg , Germany
| | - Markus Munder
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center Mainz , Mainz , Germany
| | - Ingrid Müller
- Department of Medicine, Imperial College London , London , UK
| | - Yegnasew Takele
- Department of Medicine, Imperial College London, London, UK; Leishmaniasis Research and Treatment Centre, Gondar University, Gondar, Ethiopia
| | - Pascale Kropf
- Department of Medicine, Imperial College London , London , UK
| |
Collapse
|
43
|
Okuda K, Tong M, Dempsey B, Moore KJ, Gazzinelli RT, Silverman N. Leishmania amazonensis Engages CD36 to Drive Parasitophorous Vacuole Maturation. PLoS Pathog 2016; 12:e1005669. [PMID: 27280707 PMCID: PMC4900624 DOI: 10.1371/journal.ppat.1005669] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 05/10/2016] [Indexed: 11/19/2022] Open
Abstract
Leishmania amastigotes manipulate the activity of macrophages to favor their own success. However, very little is known about the role of innate recognition and signaling triggered by amastigotes in this host-parasite interaction. In this work we developed a new infection model in adult Drosophila to take advantage of its superior genetic resources to identify novel host factors limiting Leishmania amazonensis infection. The model is based on the capacity of macrophage-like cells, plasmatocytes, to phagocytose and control the proliferation of parasites injected into adult flies. Using this model, we screened a collection of RNAi-expressing flies for anti-Leishmania defense factors. Notably, we found three CD36-like scavenger receptors that were important for defending against Leishmania infection. Mechanistic studies in mouse macrophages showed that CD36 accumulates specifically at sites where the parasite contacts the parasitophorous vacuole membrane. Furthermore, CD36-deficient macrophages were defective in the formation of the large parasitophorous vacuole typical of L. amazonensis infection, a phenotype caused by inefficient fusion with late endosomes and/or lysosomes. These data identify an unprecedented role for CD36 in the biogenesis of the parasitophorous vacuole and further highlight the utility of Drosophila as a model system for dissecting innate immune responses to infection.
Collapse
Affiliation(s)
- Kendi Okuda
- Division of Infectious Diseases & Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail: (KO); (NS)
| | - Mei Tong
- Division of Infectious Diseases & Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Brian Dempsey
- Division of Infectious Diseases & Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Kathryn J. Moore
- Department of Medicine, New York University School of Medicine, Langone Medical Center, New York, New York, United States of America
| | - Ricardo T. Gazzinelli
- Division of Infectious Diseases & Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Neal Silverman
- Division of Infectious Diseases & Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail: (KO); (NS)
| |
Collapse
|
44
|
de Menezes JP, Saraiva EM, da Rocha-Azevedo B. The site of the bite: Leishmania interaction with macrophages, neutrophils and the extracellular matrix in the dermis. Parasit Vectors 2016; 9:264. [PMID: 27146515 PMCID: PMC4857439 DOI: 10.1186/s13071-016-1540-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 04/26/2016] [Indexed: 02/07/2023] Open
Abstract
Leishmania spp., the causative agents of leishmaniasis, are intracellular parasites, transmitted to humans via the bite of their sand fly vectors. Once inoculated, the promastigotes are exposed to the dermis, which is composed of extracellular matrix (ECM), growth factors and its resident cells. Promastigote forms are phagocytosed by macrophages recruited to the site of the sand fly bite, either directly or after interaction with neutrophils. Since Leishmania is an intracellular parasite, its interaction with the host ECM has been neglected as well as the immediate steps after the sand fly bite. However, promastigotes must overcome the obstacles presented by the dermis ECM in order to establish the infection. Thus, the study of the interaction between Leishmania promastigotes and ECM components as well as the earliest stages of infection are important steps to understand the establishment of the disease, and could contribute in the future to new drug developments towards leishmaniasis.
Collapse
Affiliation(s)
| | - Elvira M Saraiva
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Bruno da Rocha-Azevedo
- Programa de Terapia Celular e Bioengenharia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil. .,Present Address: Department of Biophysics, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA.
| |
Collapse
|
45
|
Different Leishmania Species Drive Distinct Neutrophil Functions. Trends Parasitol 2016; 32:392-401. [DOI: 10.1016/j.pt.2016.02.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 02/04/2016] [Accepted: 02/08/2016] [Indexed: 01/06/2023]
|
46
|
Zhang N, Kima PE. Leishmania Infection Engages Non-Receptor Protein Kinases Differentially to Persist in Infected Hosts. Front Immunol 2016; 7:146. [PMID: 27148265 PMCID: PMC4834468 DOI: 10.3389/fimmu.2016.00146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 04/04/2016] [Indexed: 01/17/2023] Open
Abstract
Protein kinases play important roles in the regulation of cellular activities. In cells infected by pathogens, there is an increasing appreciation that dysregulated expression of protein kinases promotes the success of intracellular infections. In Leishmania-infected cells, expression and activation of protein kinases, such as the mitogen-activated protein kinases, kinases in the PI3-kinase signaling pathway, and kinases in the NF-κB-signaling pathway, are modulated in some manner. Several recent reviews have discussed our current understanding of the roles of these kinases in Leishmania infections. Apart from the kinases in the pathways enumerated above, there are other host cell protein kinases that are activated during the Leishmania infection of mammalian cells whose roles also appear to be significant. This review discusses recent observations on the Abl family of protein kinases and the protein kinase regulated by RNA in Leishmania infections.
Collapse
Affiliation(s)
- Naixin Zhang
- Department of Microbiology and Cell Science, University of Florida , Gainesville, FL , USA
| | - Peter E Kima
- Department of Microbiology and Cell Science, University of Florida , Gainesville, FL , USA
| |
Collapse
|
47
|
AKHZARI S, REZVAN H, ZOLHAVARIEH M. Expression of Pro-inflammatory Genes in Lesions and Neutrophils during Leishmania major Infection in BALB/c Mice. IRANIAN JOURNAL OF PARASITOLOGY 2016; 11:534-541. [PMID: 28127365 PMCID: PMC5251182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Leishmaniasis is a worldwide disease prevalent in tropical and sub-tropical countries in the world. Characterization of inflammatory responses produced in cutaneous leishmaniasis has not yet been completed. METHODS The specific primers were designed for ten pro-inflammatory genes including CCL4, CCL3, TNF-α, IL-1α, IL-12P35, IL-12P40, CCL5, CCR5, IL-1β and IFN- γ and their expression were assessed and compared using RT-PCR in the lesion and peripheral blood neutrophils in Leishmania infected BALB/c mice. RESULTS None of the pro-inflammatory genes was expressed in the healthy tissue and except IFN-γ others were down-regulated by the parasite in the lesion in untreated mice. In mice treated with anti-Leishmanial drugs, the expression of the pro-inflammatory genes restarted. The figure of pro-inflammatory gene expression in neutrophils was different was from the lesions in treated and untreated mice. CONCLUSION Leishmania is capable to suppress the expression of pro-inflammatory genes in the lesions but not in neutrophils. The expression of TNF-α in the lesions and down-regulation of IL-1β in neutrophils could be accounted as an indication for healing of cutaneous leishmaniasis. The results open a new window on characterization of Leishmania lesions and clarifying the role of neutrophils in Leishmania infections.
Collapse
Affiliation(s)
- Soheyla AKHZARI
- Dept. of Laboratory Sciences, School of Veterinary Sciences, Bu-Ali Sina University, Hamadan, Iran
| | - Hossein REZVAN
- Dept. of Laboratory Sciences, School of Veterinary Sciences, Bu-Ali Sina University, Hamadan, Iran,Correspondence
| | - Masoud ZOLHAVARIEH
- Dept. of Laboratory Sciences, School of Veterinary Sciences, Bu-Ali Sina University, Hamadan, Iran, Dept. of Clinical Sciences, Faculty of Para-Veterinary Sciences, Bu-Ali Sina University, Hamedan, Iran
| |
Collapse
|
48
|
Liévin-Le Moal V, Loiseau PM. Leishmania hijacking of the macrophage intracellular compartments. FEBS J 2015; 283:598-607. [PMID: 26588037 DOI: 10.1111/febs.13601] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 11/08/2015] [Accepted: 11/13/2015] [Indexed: 12/15/2022]
Abstract
Leishmania spp., transmitted to humans by the bite of the sandfly vector, are responsible for the three major forms of leishmaniasis, cutaneous, diffuse mucocutaneous and visceral. Leishmania spp. interact with membrane receptors of neutrophils and macrophages. In macrophages, the parasite is internalized within a parasitophorous vacuole and engages in a particular intracellular lifestyle in which the flagellated, motile Leishmania promastigote metacyclic form differentiates into non-motile, metacyclic amastigote form. This phenomenon is induced by Leishmania-triggered events leading to the fusion of the parasitophorous vacuole with vesicular members of the host cell endocytic pathway including recycling endosomes, late endosomes and the endoplasmic reticulum. Maturation of the parasitophorous vacuole leads to the intracellular proliferation of the Leishmania amastigote forms by acquisition of host cell nutrients while escaping host defense responses.
Collapse
Affiliation(s)
- Vanessa Liévin-Le Moal
- Anti-Parasitic Chemotherapy, Faculté de Pharmacie, CNRS, UMR 8076 BioCIS, Châtenay-Malabry, France.,Université Paris-Sud, Orsay, France.,Faculté de Pharmacie, Laboratory of Excellence in Research on Medication and Innovative Therapeutics (LabEx LERMIT), Châtenay-Malabry, France
| | - Philippe M Loiseau
- Anti-Parasitic Chemotherapy, Faculté de Pharmacie, CNRS, UMR 8076 BioCIS, Châtenay-Malabry, France.,Université Paris-Sud, Orsay, France.,Faculté de Pharmacie, Laboratory of Excellence in Research on Medication and Innovative Therapeutics (LabEx LERMIT), Châtenay-Malabry, France
| |
Collapse
|