1
|
Chaves MM. Neutrophils and purinergic signaling: Partners in the crime against Leishmania parasites? Biochimie 2025; 232:43-53. [PMID: 39855456 DOI: 10.1016/j.biochi.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 12/18/2024] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
The parasite of the genus Leishmania is the causative agent of diseases that affect humans called leishmaniasis. These diseases affect millions of people worldwide and the currently existing drugs are either very toxic or the parasites acquire resistance. Therefore, new elimination mechanisms need to be elucidated so that new therapeutic strategies can be developed. Much has already been discussed about the role of neutrophils in Leishmania infection, and their participation is still controversial. A recent study showed that receptors present in the neutrophil membrane, the purinergic receptors, can control the infection when activated, but the triggering mechanism has not been elucidated. In this review, we will address the possible participation of purinergic receptors expressed in the neutrophil extracellular membrane that may be participating in the detection of Leishmania infection and their possible effects during parasitism.
Collapse
Affiliation(s)
- Mariana M Chaves
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Bio-Manguinhos, Oswaldo Cruz Foundation, Brazilian Ministry of Health, Rio de Janeiro, Brazil.
| |
Collapse
|
2
|
Goto Y. Immunomodulation by Leishmania parasites: Potential for controlling other diseases. Parasitol Int 2025; 104:102987. [PMID: 39515578 DOI: 10.1016/j.parint.2024.102987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/01/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
In the mammalian hosts, Leishmania parasites survive and proliferate within phagolysosomes of macrophages. To avoid being killed by the immune cells, Leishmania parasites utilize their molecules to manipulate macrophages' functions for survival. Targets of such immunomodulatory molecules are not limited to macrophages, as Leishmania-derived molecules sometimes show influence on other immune cells such as neutrophils, dendritic cells, T cells and B cells. This review covers research on immunomodulation of host immunity by Leishmania parasites and introduces some examples of parasite-derived molecules participating in the immunomodulation. For example, Leishmania cell surface lipophosphoglycan (LPG) can modulate TLR2 signaling and PI3K/Akt axis in macrophages leading to induction of Th2 cells. Because chronic secretion of inflammatory cytokines is one of the causes of immune-mediated diseases such as atherosclerosis, Crohn's disease, and rheumatoid arthritis, LPG may be useful as a drug to suppress the inflammatory conditions. The unique characteristics of leishmanial molecules pose a promise as a source of immunomodulatory drugs for controlling diseases other than leishmaniasis.
Collapse
Affiliation(s)
- Yasuyuki Goto
- Laboratory of Molecular Immunology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Japan.
| |
Collapse
|
3
|
Borges-Fernandes LO, de Lima Moreira M, Pereira VHS, Pascoal-Xavier MA, Lopes Ribeiro Á, da Costa-Rocha IA, Lopes LR, Moreira GTC, Araújo MSDS, Teixeira-Carvalho A, Brito-de-Sousa JP, de Carvalho AL, Mourão MVA, Campos FA, Borges M, Carneiro M, Tsuji M, Martins-Filho OA, Coelho-dos-Reis JGA, Peruhype-Magalhães V. MR1 blockade drives differential impact on integrative signatures based on circuits of circulating immune cells and soluble mediators in visceral leishmaniasis. Front Immunol 2024; 15:1373498. [PMID: 39192975 PMCID: PMC11347828 DOI: 10.3389/fimmu.2024.1373498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/15/2024] [Indexed: 08/29/2024] Open
Abstract
Introduction Visceral leishmaniasis (VL) is an important tropical and neglected disease and represents a serious global health problem. The initial interaction between the phagocytes and the parasite is crucial to determine the pathogen's capacity to initiate infection and it shapes the subsequent immune response that will develop. While type-1 T-cells induce IL-6, IL-1β, TNF-α, and IL-12 production by monocytes/macrophages to fight the infection, type-2 T-cells are associated with a regulatory phenotype (IL-10 and TGF-β) and successful infection establishment. Recently, our group demonstrated the role of an important Th1/Th17 T-cell population, the mucosal-associated invariant T (MAIT) cells, in VL. MAIT cells can respond to L. infantum by producing TNF-α and IFN-γ upon MR1-dependent activation. Objective and methods Here, we describe the impact of the MR1-blockage on L. infantum internalization on the functional profile of circulating neutrophils and monocytes as well as the impact of the MR1-blockage on the soluble mediator signatures of in vitro whole blood cultures. Results Overall, our data showed that VL patients presents higher percentage of activated neutrophils than asymptomatic and non-infected controls. In addition, MR1 blockade led to lower TNF-α and TGF-β production by non-activated neutrophils from asymptomatic individuals. Moreover, TNF-α and IL-10 production by monocytes was higher in VL patients. In the analysis of soluble mediators produced in vitro, MR1-blockade induced a decrease of IFN-γ and an increase of IL-10, IL-27 and IL-33 in the cell cultures of AS group, a cytokine pattern associated with type 2 deleterious response. Discussion and conclusion These data corroborate the hypothesis that MR1-restricted responses are associated to a protective role during Leishmania infection.
Collapse
Affiliation(s)
| | - Marcela de Lima Moreira
- René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ-MINAS), Belo Horizonte, Minas Gerais, Brazil
| | | | - Marcelo Antônio Pascoal-Xavier
- René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ-MINAS), Belo Horizonte, Minas Gerais, Brazil
- School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ágata Lopes Ribeiro
- René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ-MINAS), Belo Horizonte, Minas Gerais, Brazil
- Basic and Applied Virology Laboratory, Department of Microbiology, Institute for Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Ludmila Rosa Lopes
- René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ-MINAS), Belo Horizonte, Minas Gerais, Brazil
| | | | | | - Andréa Teixeira-Carvalho
- René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ-MINAS), Belo Horizonte, Minas Gerais, Brazil
| | | | - Andrea Lucchesi de Carvalho
- João Paulo II Children’s Hospital, Fundação Hospitalar do Estado de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Flávia Alves Campos
- João Paulo II Children’s Hospital, Fundação Hospitalar do Estado de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marineide Borges
- João Paulo II Children’s Hospital, Fundação Hospitalar do Estado de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mariângela Carneiro
- Parasitology Department, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Moriya Tsuji
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
- Division of Infectious Disease, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | | | - Jordana Grazziela Alves Coelho-dos-Reis
- René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ-MINAS), Belo Horizonte, Minas Gerais, Brazil
- Basic and Applied Virology Laboratory, Department of Microbiology, Institute for Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Rezaei Z, Tahmasebi A, Pourabbas B. Using meta-analysis and machine learning to investigate the transcriptional response of immune cells to Leishmania infection. PLoS Negl Trop Dis 2024; 18:e0011892. [PMID: 38190401 PMCID: PMC10798641 DOI: 10.1371/journal.pntd.0011892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 01/19/2024] [Accepted: 12/29/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND Leishmaniasis is a parasitic disease caused by the Leishmania protozoan affecting millions of people worldwide, especially in tropical and subtropical regions. The immune response involves the activation of various cells to eliminate the infection. Understanding the complex interplay between Leishmania and the host immune system is crucial for developing effective treatments against this disease. METHODS This study collected extensive transcriptomic data from macrophages, dendritic, and NK cells exposed to Leishmania spp. Our objective was to determine the Leishmania-responsive genes in immune system cells by applying meta-analysis and feature selection algorithms, followed by co-expression analysis. RESULTS As a result of meta-analysis, we discovered 703 differentially expressed genes (DEGs), primarily associated with the immune system and cellular metabolic processes. In addition, we have substantiated the significance of transcription factor families, such as bZIP and C2H2 ZF, in response to Leishmania infection. Furthermore, the feature selection techniques revealed the potential of two genes, namely G0S2 and CXCL8, as biomarkers and therapeutic targets for Leishmania infection. Lastly, our co-expression analysis has unveiled seven hub genes, including PFKFB3, DIAPH1, BSG, BIRC3, GOT2, EIF3H, and ATF3, chiefly related to signaling pathways. CONCLUSIONS These findings provide valuable insights into the molecular mechanisms underlying the response of immune system cells to Leishmania infection and offer novel potential targets for the therapeutic goals.
Collapse
Affiliation(s)
- Zahra Rezaei
- Professor Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Tahmasebi
- Professor Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahman Pourabbas
- Professor Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
7
|
Kumar R, Bhatia M, Pai K. Role of Chemokines in the Pathogenesis of Visceral Leishmaniasis. Curr Med Chem 2022; 29:5441-5461. [PMID: 35579167 DOI: 10.2174/0929867329666220509171244] [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: 08/04/2021] [Revised: 12/23/2021] [Accepted: 03/02/2022] [Indexed: 11/22/2022]
Abstract
Visceral leishmaniasis (VL; also known as kala-azar), caused by the protozoan parasite Leishmania donovani, is characterized by the inability of the host to generate an effective immune response. The manifestations of the disease depend on the involvement of various immune components such as activation of macrophages, cell mediated immunity, secretion of cytokines and chemokines, etc. Macrophages are the final host cells for Leishmania parasites to multiply, and they are the key to a controlled or aggravated response that leads to clinical symptoms. The two most common macrophage phenotypes are M1 and M2. The pro-inflammatory microenvironment (mainly by IL-1β, IL-6, IL-12, IL-23, and TNF-α cytokines) and tissue injury driven by classically activated macrophages (M1-like) and wound healing driven by alternatively activated macrophages (M2-like) in an anti-inflammatory environment (mainly by IL-10, TGF-β, chemokine ligand (CCL)1, CCL2, CCL17, CCL18, and CCL22). Moreover, on polarized Th cells, chemokine receptors are expressed differently. Typically, CXCR3 and CCR5 are preferentially expressed on polarized Th1 cells, whereas CCR3, CCR4, and CCR8 have been associated with the Th2 phenotype. Further, the ability of the host to produce a cell-mediated immune response capable of regulating and/or eliminating the parasite is critical in the fight against the disease. Here, we review the interactions between parasites and chemokines and chemokine receptors in the pathogenesis of VL.
Collapse
Affiliation(s)
- Ramesh Kumar
- Department of Biochemistry, Bundelkhand University, Jhansi (UP), India
| | - Madhav Bhatia
- Department of Pathology and Biomedical Sciences, University of Otago, Christchurch, New Zealand
| | - Kalpana Pai
- Department of Zoology, Savitribai Phule Pune University, Pune, Maharashtra, India
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Varikuti S, Verma C, Holcomb E, Jha BK, Viana A, Maryala R, Lamenza F, McElwain BK, Doni NY, Papenfuss T, Oghumu S, Gannavaram S, Nakhasi HL, Satoskar AR. MicroRNA-21 Deficiency Promotes the Early Th1 Immune Response and Resistance toward Visceral Leishmaniasis. THE JOURNAL OF IMMUNOLOGY 2021; 207:1322-1332. [PMID: 34341171 DOI: 10.4049/jimmunol.2001099] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 06/03/2021] [Indexed: 12/15/2022]
Abstract
MicroRNA-21 (miR-21) inhibits IL-12 expression and impairs the Th1 response necessary for control of Leishmania infection. Recent studies have shown that Leishmania infection induces miR-21 expression in dendritic cells and macrophages, and inhibition of miR-21 restores IL-12 expression. Because miR-21 is known to be expressed due to inflammatory stimuli in a wide range of hematopoietic cells, we investigated the role of miR-21 in regulating immune responses during visceral leishmaniasis (VL) caused by Leishmania donovani infection. We found that miR-21 expression was significantly elevated in dendritic cells, macrophages, inflammatory monocytes, polymorphonuclear neutrophils, and in the spleen and liver tissues after L. donovani infection, concomitant with an increased expression of disease exacerbating IL-6 and STAT3. Bone marrow dendritic cells from miR-21 knockout (miR-21KO) mice showed increased IL-12 production and decreased production of IL-10. On L. donovani infection, miR-21KO mice exhibited significantly greater numbers of IFN-γ- and TNF-α-producing CD4+ and CD8+ T cells in their organs that was associated with increased production of Th1-associated IFN-γ, TNF-α, and NO from the splenocytes. Finally, miR-21KO mice displayed significantly more developing and mature hepatic granulomas leading to reduction in organ parasitic loads compared with wild type counterparts. Similar results were noted in L. donovani-infected wild type mice after transient miR-21 depletion. These observations indicate that miR-21 plays a critical role in pathogenesis of VL by suppressing IL-12- and Th1-associated IFN-γ and also inducing disease-promoting induction of the IL-6 and STAT-3 signaling pathway. miR-21 could therefore be used as a potential target for developing host-directed treatment for VL.
Collapse
Affiliation(s)
- Sanjay Varikuti
- Department of Pathology, The Ohio State University Medical Center, Columbus, OH.,Department of Bioscience and Biotechnology, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Chaitenya Verma
- Department of Pathology, The Ohio State University Medical Center, Columbus, OH
| | - Erin Holcomb
- Department of Pathology, The Ohio State University Medical Center, Columbus, OH
| | - Bijay Kumar Jha
- Division of Infectious Diseases, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Agostinho Viana
- Department of Pathology, The Ohio State University Medical Center, Columbus, OH
| | - Ritvik Maryala
- Department of Pathology, The Ohio State University Medical Center, Columbus, OH
| | - Felipe Lamenza
- Department of Pathology, The Ohio State University Medical Center, Columbus, OH
| | - Bryan K McElwain
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Nebiye Yentur Doni
- Department of Pathology, The Ohio State University Medical Center, Columbus, OH
| | - Tracey Papenfuss
- College of Veterinary Medicine, The Ohio State University, Columbus, OH
| | - Steve Oghumu
- Department of Pathology, The Ohio State University Medical Center, Columbus, OH
| | - Sreenivas Gannavaram
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD; and
| | - Hira L Nakhasi
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD; and
| | - Abhay R Satoskar
- Department of Pathology, The Ohio State University Medical Center, Columbus, OH; .,Department of Microbiology, The Ohio State University, Columbus, OH
| |
Collapse
|
11
|
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
|
12
|
Chaves MM, Savio LEB, Coutinho-Silva R. Purinergic signaling: a new front-line determinant of resistance and susceptibility in leishmaniasis. Biomed J 2021; 45:109-117. [PMID: 34175493 PMCID: PMC9133308 DOI: 10.1016/j.bj.2021.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 12/12/2022] Open
Abstract
Leishmaniasis is a neglected tropical disease that causes several clinical manifestations. Parasites of the genus Leishmania cause this disease. Spread across five continents, leishmaniasis is a particular public health problem in developing countries. Leishmania infects phagocytic cells such as macrophages, where it induces adenosine triphosphate (ATP) release at the time of infection. ATP activates purinergic receptors in the cell membranes of infected cells and promotes parasite control by inducing leukotriene B4 release and NLRP3 inflammasome activation. Moreover, uridine triphosphate induces ATP release, exacerbating the immune response. However, ATP may also undergo catalysis by ectonucleotidases present in the parasite membrane, generating adenosine, which activates P1 receptors and induces the production of anti-inflammatory molecules such as prostaglandin E2 and IL-10. These mechanisms culminate in Leishmania's survival. Thus, how Leishmania handles extracellular nucleotides and the activation of purinergic receptors determines the control or the dissemination of the disease.
Collapse
Affiliation(s)
- Mariana M Chaves
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Luiz Eduardo B Savio
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Robson Coutinho-Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
13
|
Passelli K, Billion O, Tacchini-Cottier F. The Impact of Neutrophil Recruitment to the Skin on the Pathology Induced by Leishmania Infection. Front Immunol 2021; 12:649348. [PMID: 33732265 PMCID: PMC7957080 DOI: 10.3389/fimmu.2021.649348] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/04/2021] [Indexed: 12/29/2022] Open
Abstract
Leishmania (L.) are obligate intracellular protozoan parasites that cause the leishmaniases, a spectrum of neglected infectious vector-borne diseases with a broad range of clinical manifestations ranging from local cutaneous, to visceral forms of the diseases. The parasites are deposited in the mammalian skin during the blood meal of an infected female phlebotomine sand fly. The skin is a complex organ acting as the first line of physical and immune defense against pathogens. Insults to skin integrity, such as that occurring during insect feeding, induces the local secretion of pro-inflammatory molecules generating the rapid recruitment of neutrophils. At the site of infection, skin keratinocytes play a first role in host defense contributing to the recruitment of inflammatory cells to the infected dermis, of which neutrophils are the first recruited cells. Although neutrophils efficiently kill various pathogens including Leishmania, several Leishmania species have developed mechanisms to survive in these cells. In addition, through their rapid release of cytokines, neutrophils modulate the skin microenvironment at the site of infection, a process shaping the subsequent development of the adaptive immune response. Neutrophils may also be recruited later on in unhealing forms of cutaneous leishmaniasis and to the spleen and liver in visceral forms of the disease. Here, we will review the mechanisms involved in neutrophil recruitment to the skin following Leishmania infection focusing on the role of keratinocytes in this process. We will also discuss the distinct involvement of neutrophils in the outcome of leishmaniasis.
Collapse
Affiliation(s)
- Katiuska Passelli
- Department of Biochemistry, WHO Collaborative Centre for Research and Training in Immunology, University of Lausanne, Lausanne, Switzerland
| | - Oaklyne Billion
- Department of Biochemistry, WHO Collaborative Centre for Research and Training in Immunology, University of Lausanne, Lausanne, Switzerland
| | - Fabienne Tacchini-Cottier
- Department of Biochemistry, WHO Collaborative Centre for Research and Training in Immunology, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
14
|
Samant M, Sahu U, Pandey SC, Khare P. Role of Cytokines in Experimental and Human Visceral Leishmaniasis. Front Cell Infect Microbiol 2021; 11:624009. [PMID: 33680991 PMCID: PMC7930837 DOI: 10.3389/fcimb.2021.624009] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/22/2021] [Indexed: 12/16/2022] Open
Abstract
Visceral Leishmaniasis (VL) is the most fatal form of disease leishmaniasis. To date, there are no effective prophylactic measures and therapeutics available against VL. Recently, new immunotherapy-based approaches have been established for the management of VL. Cytokines, which are predominantly produced by helper T cells (Th) and macrophages, have received great attention that could be an effective immunotherapeutic approach for the treatment of human VL. Cytokines play a key role in forming the host immune response and in managing the formation of protective and non-protective immunities during infection. Furthermore, immune response mediated through different cytokines varies from different host or animal models. Various cytokines viz. IFN-γ, IL-2, IL-12, and TNF-α play an important role during protection, while some other cytokines viz. IL-10, IL-6, IL-17, TGF-β, and others are associated with disease progression. Therefore, comprehensive knowledge of cytokine response and their interaction with various immune cells is very crucial to determine appropriate immunotherapies for VL. Here, we have discussed the role of cytokines involved in VL disease progression or host protection in different animal models and humans that will determine the clinical outcome of VL and open the path for the development of rapid and accurate diagnostic tools as well as therapeutic interventions against VL.
Collapse
Affiliation(s)
- Mukesh Samant
- Cell and Molecular Biology Laboratory, Department of Zoology, Kumaun University, Almora, India
| | - Utkarsha Sahu
- Department of Microbiology, All India Institute of Medical Sciences, Bhopal, India
| | - Satish Chandra Pandey
- Cell and Molecular Biology Laboratory, Department of Zoology, Kumaun University, Almora, India
| | - Prashant Khare
- Department of Microbiology, All India Institute of Medical Sciences, Bhopal, India
| |
Collapse
|
15
|
Abdullahi IN, Emeribe AU, Adekola HA, Muhammad HY, Ahmad AEF, Anka AU, Mohammed Y, Haruna S, Oderinde BS, Shuwa HA, Babayo A. Leucocytes and Th-associated Cytokine Profile of HIV-Leishmaniasis Co-Infected Persons Attending Abuja Teaching Hospital, Nigeria. Eurasian J Med 2020; 52:271-276. [PMID: 33209080 DOI: 10.5152/eurasianjmed.2020.20008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Objective T-helper cells (Th)-1& -2 cytokines homeostasis control or predict clinical outcome of infected persons, especially those with HIV /AIDS. This case-control study evaluated the leucocytes differentials, TNF-alpha, interleukin (IL)-2 and -10 levels among HIV infected persons with serological evidence of leishmaniasis attending University of Abuja Teaching Hospital, Nigeria. Materials and Methods Blood samples from 28 HIV infected persons who had Leishmania donovani rK39 and Immunoglobulin-G (IgG) positive (group 1), 30 age- & -sex matched HIV infected persons without Leishmania antibodies (group 2) and 30 apparently healthy persons without HIV and Leishmania antibodies (group 3). Full blood counts, TNF alpha, IL-2 and -10 levels were analyzed using automated hematology analyzer and ELISA, respectively. Structured questionnaires were used to collate biodata and clinical presentations of participants. Results Ten (35.7%) participants in group 1 were on ART, 15 (50%) in group 2 were on ART, while group 3 were ART naïve. There were significantly higher values in basophil (4.4±2.5%) and eosinophil counts (12.9±3.8%) in HIV/leishmania coinfected persons (p<0.005). However, other white cells subpopulation was significantly lower in HIV/leishmania co-infected participants (p<0.05). There was significantly reduced CD4+ T cell counts ([119±26 versus 348±63 versus 605±116 cells/mm3]), TNF-alpha ([36.82±8.21 versus 64.67±12.54 versus 254.98±65.59 pg/mL]) and IL-2 levels ([142.14±20.91 versus 507.6±84.42 versus 486.62±167.87 pg/mL]) among HIV/Leishmania co-infected participants compared to group 2 and group 3 participants, respectively. However, higher IL-10 level (80.35±14.57 pg/mL) was found in HIV/Leishmania co-infected participants as opposed to the HIV monoinfected (62.2±10.43 pg/mL) and apparently healthy persons (23.97±4.88 pg/mL) (p<0.001). Conclusion Eosinophil, basophil counts and serum IL-10 level were high in HIV/Leishmania coinfected persons, demonstrating parasite-induced hypersensitivity and immunosuppression.
Collapse
Affiliation(s)
- Idris Nasir Abdullahi
- Department of Medical Laboratory Science, Faculty of Allied Health Sciences, Ahmadu Bello University, Zaria, Nigeria
| | | | | | | | - Abdurrahman El-Fulaty Ahmad
- Department of Medical Laboratory Science, Faculty of Allied Health Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Abubakar Umar Anka
- Department of Medical Laboratory Science, Faculty of Allied Health Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Yusuf Mohammed
- Department of Medical Microbiology and Parasitology, Bayero University, Kano, Nigeria
| | - Shamsuddeen Haruna
- Department of Medical Laboratory Science, Faculty of Allied Health Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Bamidele Soji Oderinde
- Department of Medical Laboratory Science, Faculty of Allied Health Sciences, University of Maiduguri, Maiduguri, Nigeria
| | - Halima Ali Shuwa
- Department of Community Health, Federal University Dutse, Jigawa State, Nigeria
| | - Adamu Babayo
- Department of Medical Microbiology and Parasitology, Bayero University, Kano, Nigeria
| |
Collapse
|
16
|
Bhattacharya P, Dey R, Saxena A, Karmakar S, Ismail N, Gannavaram S, Dagur PK, Satoskar M, Satoskar S, De Paoli S, Takeda K, McCoy JP, Nakhasi HL. Essential Role of Neutrophils in the Protective Immune Response Induced by a Live Attenuated Leishmania Vaccine. THE JOURNAL OF IMMUNOLOGY 2020; 205:3333-3347. [PMID: 33177159 DOI: 10.4049/jimmunol.2000829] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/01/2020] [Indexed: 12/13/2022]
Abstract
No licensed vaccine exists against visceral leishmaniasis (VL), a disease caused by the Leishmania donovani parasite. We have previously reported both macrophages and dendritic cells play important role in the protection induced by a live attenuated centrin gene-deleted L. donovani (LdCen-/- ) parasite vaccine. The role of neutrophils in orchestrating the initial innate response to pathogens is widely recognized. To investigate the early interaction of LdCen-/- with neutrophils, we immunized mice intradermally in the ear pinna with LdCen-/- Compared with LdWT infection, LdCen-/- parasites induced higher recruitment of neutrophils to the ear dermis and ear draining lymph nodes (dLN) as early as 6-18 h after immunization, which were predominantly proinflammatory in nature. Neutrophils from ear dLN of LdCen-/- -immunized mice exhibited heightened expression of costimulatory molecules and attenuated expression of coinhibitory molecules necessary for higher T cell activation. Further phenotypic characterization revealed heterogeneous neutrophil populations containing Nα and Nβ subtypes in the ear dLN. Of the two, the parasitized Nα subset from LdCen-/- -immunized mice exhibited much stronger Ag-specific CD4+ T cell proliferation ex vivo. Adoptive transfer of neutrophils bearing LdCen-/- parasites induced an increased Th1 response in naive mice. Importantly, neutrophil depletion significantly abrogated Ag-specific CD4+ T cell proliferation in LdCen-/- -immunized mice and impaired protection against virulent challenge. Conversely, replenishing of neutrophils significantly restored the LdCen-/- -induced host-protective response. These results suggest that neutrophils are indispensable for protective immunity induced by LdCen-/- parasite vaccine.
Collapse
Affiliation(s)
- Parna Bhattacharya
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993;
| | - Ranadhir Dey
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993
| | - Ankit Saxena
- Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Subir Karmakar
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993
| | - Nevien Ismail
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993
| | - Sreenivas Gannavaram
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993
| | - Pradeep K Dagur
- Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | | | | | - Silvia De Paoli
- Office of Blood Research and Review, U.S. Food and Drug Administration, Silver Spring, MD 20993; and
| | - Kazuyo Takeda
- Microscopy and Imaging Core Facility, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993
| | - John Philip McCoy
- Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Hira L Nakhasi
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993;
| |
Collapse
|
17
|
Jafarzadeh A, Nemati M, Sharifi I, Nair A, Shukla D, Chauhan P, Khorramdelazad H, Sarkar A, Saha B. Leishmania species-dependent functional duality of toll-like receptor 2. IUBMB Life 2019; 71:1685-1700. [PMID: 31329370 DOI: 10.1002/iub.2129] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 06/25/2019] [Indexed: 01/15/2023]
Abstract
Toll-like receptors (TLRs) are a subset of pattern recognition receptors (PRR) in innate immunity and act as a connecting link between innate and adaptive immune systems. During Leishmania infection, the activation of TLRs influences the pathogen-specific immune responses, which may play a decisive role in determining the outcome of infection, toward elimination or survival of the pathogen. Antigen-presenting cells (APCs) of the innate immune system such as macrophages, dendritic cells (DCs), neutrophils, natural killer (NK) cells, and NKT cells express TLR2, which plays a crucial role in the parasite recognition and elicitation of immune responses in Leishmania infection. Depending on the infecting Leishmania species, the TLR2 pathways may result in a host-protective or a disease-exacerbating response. While Leishmania major and Leishmania donovani infections trigger TLR2-related host-protective and non-protective immune responses, Leishmania mexicana and Leishmania infantum infections are reported to elicit TLR2-mediated host-protective responses and Leishmania amazonensis and Leishmania braziliensis infections are reported to evoke a disease-exacerbating response. These findings illustrate that TLR2-related effector functions are diverse and may be exerted in a species- or strain-dependent manner. TLR2 agonists or antagonists may have therapeutic potentials to trigger the desired immune response during leishmaniasis. In this review, we discuss the TLR2-related immune responses during leishmaniasis and highlight the novel insights into the possible role of TLR2-driven resistance or susceptibility to Leishmania.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.,Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Maryam Nemati
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.,Department of Haematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Iraj Sharifi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Arathi Nair
- National Centre for Cell Science, Pune, India
| | | | | | - Hossain Khorramdelazad
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Arup Sarkar
- Trident Academy of Creative Technology, Bhubaneswar, India
| | - Bhaskar Saha
- National Centre for Cell Science, Pune, India.,Trident Academy of Creative Technology, Bhubaneswar, India
| |
Collapse
|
18
|
Leishmania major degrades murine CXCL1 - An immune evasion strategy. PLoS Negl Trop Dis 2019; 13:e0007533. [PMID: 31260451 PMCID: PMC6625741 DOI: 10.1371/journal.pntd.0007533] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 07/12/2019] [Accepted: 06/07/2019] [Indexed: 12/19/2022] Open
Abstract
Leishmaniasis is a global health problem with an estimated report of 2 million new cases every year and more than 1 billion people at risk of contracting this disease in endemic areas. The innate immune system plays a central role in controlling L. major infection by initiating a signaling cascade that results in production of pro-inflammatory cytokines and recruitment of both innate and adaptive immune cells. Upon infection with L. major, CXCL1 is produced locally and plays an important role in the recruitment of neutrophils to the site of infection. Herein, we report that L. major specifically targets murine CXCL1 for degradation. The degradation of CXCL1 is not dependent on host factors as L. major can directly degrade recombinant CXCL1 in a cell-free system. Using mass spectrometry, we discovered that the L. major protease cleaves at the C-terminal end of murine CXCL1. Finally, our data suggest that L. major metalloproteases are involved in the direct cleavage and degradation of CXCL1, and a synthetic peptide spanning the CXCL1 cleavage site can be used to inhibit L. major metalloprotease activity. In conclusion, our study has identified an immune evasion strategy employed by L. major to evade innate immune responses in mice, likely reservoirs in the endemic areas, and further highlights that targeting these L. major metalloproteases may be important in controlling infection within the reservoir population and transmittance of the disease. Our study discovered a highly specific role for L. major metalloprotease in cleaving and degrading murine CXCL1. Indeed, L. major metalloprotease did not cleave murine CXCL2 or human CXCL1, CXCL2 and CXCL8. CXCL1 is a critical chemokine required for neutrophil recruitment to the site of infection; thus, we propose that this metalloprotease may have evolved to evade immune responses specifically in the murine host. We have further identified that the C-terminal end on CXCL1 is targeted for cleavage by the L. major metalloprotease. Finally, this cleavage site information was used to design peptides that are able to inhibit CXCL1 degradation by L. major. Our study highlights an immune evasion strategy utilized by L. major to establish infection within a murine host.
Collapse
|
19
|
Schroeder J, Ross K, McIntosh K, Jabber S, Woods S, Crowe J, Patterson Kane J, Alexander J, Lawrence C, Plevin R. Novel protective role for MAP kinase phosphatase 2 in inflammatory arthritis. RMD Open 2019; 5:e000711. [PMID: 30713718 PMCID: PMC6340532 DOI: 10.1136/rmdopen-2018-000711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 11/21/2018] [Accepted: 11/25/2018] [Indexed: 12/14/2022] Open
Abstract
Objectives We have previously shown mitogen-activated protein kinase phosphatase 2 (MKP-2) to be a key regulator of proinflammatory cytokines in macrophages. In the study presented here, we investigated the role of MKP-2 in inflammatory arthritis with a particular focus on neutrophils. Methods To achieve this, we subjected MKP-2 deficient and wild type mice to collagen antibody induced arthritis, an innate model of arthritis, and determined disease pathology. To further our investigation, we depleted neutrophils in a prophylactic and therapeutic fashion. Last, we used chemotaxis assays to analyse the impact of MKP-2 deletion on neutrophil migration. Results MKP-2-/- mice showed a significant increase in disease pathology linked to elevated levels of proarthritic cytokines and chemokines TNF-α, IL-6 and MCP-1 in comparison to wild type controls. This phenotype is prevented or abolished after administration of neutrophil depleting antibody prior or after onset of disease, respectively. While MCP-1 levels were not affected, neutrophil depletion diminished TNF-α and reduced IL-6, thus linking these cytokines to neutrophils. In vivo imaging showed that MKP-2-/- mice had an increased influx of neutrophils into affected joints, which was higher and potentially prolonged than in wild type animals. Furthermore, using chemotaxis assays we revealed that MKP-2 deficient neutrophils migrate faster towards a Leukotriene B4 gradient. This process correlated with a reduced phosphorylation of ERK in MKP-2-/- neutrophils. Conclusions This is the first study to show a protective role for MKP-2 in inflammatory arthritis.
Collapse
Affiliation(s)
- Juliane Schroeder
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, Scotland
| | - Kirsty Ross
- Pure and Applied Chemistry, Technology Innovation Centre, University of Strathclyde, Glasgow, Scotland
| | - Kathryn McIntosh
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland
| | - Shilan Jabber
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland
| | - Stuart Woods
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland
| | - Jenny Crowe
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, Scotland
| | | | - James Alexander
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland
| | - Catherine Lawrence
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland
| | - Robin Plevin
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland
| |
Collapse
|
20
|
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
|
21
|
Regli IB, Fernández OL, Martínez-Salazar B, Gómez MA, Saravia NG, Tacchini-Cottier F. Resistance of Leishmania (Viannia) Panamensis to Meglumine Antimoniate or Miltefosine Modulates Neutrophil Effector Functions. Front Immunol 2018; 9:3040. [PMID: 30622537 PMCID: PMC6308327 DOI: 10.3389/fimmu.2018.03040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/10/2018] [Indexed: 01/16/2023] Open
Abstract
Leishmania (Viannia) panamensis (L. (V.) p.) is the main causative agent of cutaneous leishmaniasis in Colombia and is usually treated with either meglumine antimoniate (MA) or miltefosine (MIL). In recent years, there has been increasing evidence of the emergence of drug-resistance against these compounds. Neutrophils are known to play an important role in immunity against Leishmania. These cells are rapidly recruited upon infection and are also present in chronic lesions. However, their involvement in the outcome of infection with drug-resistant Leishmania has not been examined. In this study, human and murine neutrophils were infected in vitro with MA or MIL drug-resistant L. (V.) p. lines derived from a parental L. (V.) p. drug-susceptible strain. Neutrophil effector functions were assessed analyzing the production of reactive oxygen species (ROS), the formation of neutrophil extracellular trap (NET) and the expression of cell surface activation markers. Parasite killing by neutrophils was assessed using L. (V.) p. transfected with a luciferase reporter. We show here that MA and MIL-resistant L. (V.) p. lines elicited significantly increased NET formation and MA-resistant L. (V.) p. induced significantly increased ROS production in both murine and human neutrophils, compared to infections with the parental MIL and MA susceptible strain. Furthermore, neutrophils exposed to drug-resistant lines showed increased activation, as revealed by decreased expression of CD62L and increased expression of CD66b in human neutrophils yet presented higher survival within neutrophils than the drug-susceptible strain. These results provide evidence that parasite drug-susceptibility may influences neutrophil activation and function as well as parasite survival within neutrophils. Further investigaton of the inter-relationship of drug susceptibility and neutrophil effector function should contribute to better understanding of the factors involved in susceptibility to anti-Leishmania drugs.
Collapse
Affiliation(s)
- Ivo B Regli
- Department of Biochemistry, WHO-Immunology Research and Training Center, University of Lausanne, Epalinges, Switzerland
| | - Olga Lucía Fernández
- Centro Internacional de Entrenamiento e Investigaciones Médicas, Cali, Colombia.,CIDEIM, Universidad ICESI, Cali, Colombia
| | - Berenice Martínez-Salazar
- Department of Biochemistry, WHO-Immunology Research and Training Center, University of Lausanne, Epalinges, Switzerland
| | - Maria Adelaida Gómez
- Centro Internacional de Entrenamiento e Investigaciones Médicas, Cali, Colombia.,CIDEIM, Universidad ICESI, Cali, Colombia
| | - Nancy Gore Saravia
- Centro Internacional de Entrenamiento e Investigaciones Médicas, Cali, Colombia.,CIDEIM, Universidad ICESI, Cali, Colombia
| | - Fabienne Tacchini-Cottier
- Department of Biochemistry, WHO-Immunology Research and Training Center, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
22
|
Wun K, Theriault BR, Pierre JF, Chen EB, Leone VA, Harris KG, Xiong L, Jiang Q, Spedale M, Eskandari OM, Chang EB, Ho KJ. Microbiota control acute arterial inflammation and neointimal hyperplasia development after arterial injury. PLoS One 2018; 13:e0208426. [PMID: 30521585 PMCID: PMC6283560 DOI: 10.1371/journal.pone.0208426] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 11/16/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The microbiome has a functional role in a number of inflammatory processes and disease states. While neointimal hyperplasia development has been linked to inflammation, a direct role of the microbiota in neointimal hyperplasia has not yet been established. Germ-free (GF) mice are an invaluable model for studying causative links between commensal organisms and the host. We hypothesized that GF mice would exhibit altered neointimal hyperplasia following carotid ligation compared to conventionally raised (CONV-R) mice. METHODS Twenty-week-old male C57BL/6 GF mice underwent left carotid ligation under sterile conditions. Maintenance of sterility was assessed by cultivation and 16S rRNA qPCR of stool. Neointimal hyperplasia was assessed by morphometric and histologic analysis of arterial sections after 28 days. Local arterial cell proliferation and inflammation was assessed by immunofluorescence for Ki67 and inflammatory cell markers at five days. Systemic inflammation was assessed by multiplex immunoassays of serum. CONV-R mice treated in the same manner served as the control cohort. GF and CONV-R mice were compared using standard statistical methods. RESULTS All GF mice remained sterile during the entire study period. Twenty-eight days after carotid ligation, CONV-R mice had significantly more neointimal hyperplasia development compared to GF mice, as assessed by intima area, media area, intima+media area, and intima area/(intima+media) area. The collagen content of the neointimal lesions appeared qualitatively similar on Masson's trichrome staining. There was significantly reduced Ki67 immunoreactivity in the media and adventitia of GF carotid arteries 5 days after ligation. GF mice also had increased arterial infiltration of anti-inflammatory M2 macrophages compared to CONV-R mouse arteries and a reduced proportion of mature neutrophils. GF mice had significantly reduced serum IFN-γ-inducible protein (IP)-10 and MIP-2 5 days after carotid ligation, suggesting a reduced systemic inflammatory response. CONCLUSIONS GF mice have attenuated neointimal hyperplasia development compared to CONV-R mice, which is likely related to altered kinetics of wound healing and acute inflammation. Recognizing the role of commensals in the regulation of arterial remodeling will provide a deeper understanding of the pathophysiology of restenosis and support strategies to treat or reduce restenosis risk by manipulating microbiota.
Collapse
Affiliation(s)
- Kelly Wun
- Division of Vascular Surgery, Northwestern University, Chicago, IL, United States of America
| | - Betty R. Theriault
- Department of Surgery and Animal Resources Center, University of Chicago, Chicago, IL, United States of America
| | - Joseph F. Pierre
- Department of Pediatrics, The University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Edmund B. Chen
- Division of Vascular Surgery, Northwestern University, Chicago, IL, United States of America
| | - Vanessa A. Leone
- Department of Medicine, Section of Gastroenterology, University of Chicago, Chicago, IL, United States of America
| | - Katharine G. Harris
- Department of Medicine, Section of Gastroenterology, University of Chicago, Chicago, IL, United States of America
| | - Liqun Xiong
- Division of Vascular Surgery, Northwestern University, Chicago, IL, United States of America
| | - Qun Jiang
- Division of Vascular Surgery, Northwestern University, Chicago, IL, United States of America
| | - Melanie Spedale
- Department of Surgery and Animal Resources Center, University of Chicago, Chicago, IL, United States of America
| | - Owen M. Eskandari
- Division of Vascular Surgery, Northwestern University, Chicago, IL, United States of America
| | - Eugene B. Chang
- Department of Medicine, Section of Gastroenterology, University of Chicago, Chicago, IL, United States of America
| | - Karen J. Ho
- Division of Vascular Surgery, Northwestern University, Chicago, IL, United States of America
| |
Collapse
|
23
|
Tomiotto-Pellissier F, Bortoleti BTDS, Assolini JP, Gonçalves MD, Carloto ACM, Miranda-Sapla MM, Conchon-Costa I, Bordignon J, Pavanelli WR. Macrophage Polarization in Leishmaniasis: Broadening Horizons. Front Immunol 2018; 9:2529. [PMID: 30429856 PMCID: PMC6220043 DOI: 10.3389/fimmu.2018.02529] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/15/2018] [Indexed: 01/14/2023] Open
Abstract
Leishmaniasis is a vector-borne neglected tropical disease that affects more than 700,000 people annually. Leishmania parasites cause the disease, and different species trigger a distinct immune response and clinical manifestations. Macrophages are the final host cells for the proliferation of Leishmania parasites, and these cells are the key to a controlled or exacerbated response that culminates in clinical manifestations. M1 and M2 are the two main macrophage phenotypes. M1 is a pro-inflammatory subtype with microbicidal properties, and M2, or alternatively activated, is an anti-inflammatory/regulatory subtype that is related to inflammation resolution and tissue repair. The present review elucidates the roles of M1 and M2 polarization in leishmaniasis and highlights the role of the salivary components of the vector and the action of the parasite in the macrophage plasticity.
Collapse
Affiliation(s)
- Fernanda Tomiotto-Pellissier
- Biosciences and Biotechnology Postgraduate Program, Carlos Chagas Institute (ICC), Fiocruz, Curitiba, Brazil.,Laboratory of Immunoparasitology, Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - Bruna Taciane da Silva Bortoleti
- Biosciences and Biotechnology Postgraduate Program, Carlos Chagas Institute (ICC), Fiocruz, Curitiba, Brazil.,Laboratory of Immunoparasitology, Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - João Paulo Assolini
- Laboratory of Immunoparasitology, Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - Manoela Daiele Gonçalves
- Laboratory of Biotransformation and Phytochemistry, Department of Chemistry, State University of Londrina, Universitary Hospital, Londrina, Brazil
| | | | | | - Ivete Conchon-Costa
- Laboratory of Immunoparasitology, Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - Juliano Bordignon
- Biosciences and Biotechnology Postgraduate Program, Carlos Chagas Institute (ICC), Fiocruz, Curitiba, Brazil.,Laboratory of Molecular Virology, Carlos Chagas Institute (ICC), Fiocruz, Curitiba, Brazil
| | - Wander Rogério Pavanelli
- Biosciences and Biotechnology Postgraduate Program, Carlos Chagas Institute (ICC), Fiocruz, Curitiba, Brazil.,Laboratory of Immunoparasitology, Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| |
Collapse
|
24
|
Barbosa LA, Fiuza PP, Borges LJ, Rolim FA, Andrade MB, Luz NF, Quintela-Carvalho G, Lima JB, Almeida RP, Chan FK, Bozza MT, Borges VM, Prates DB. RIPK1-RIPK3-MLKL-Associated Necroptosis Drives Leishmania infantum Killing in Neutrophils. Front Immunol 2018; 9:1818. [PMID: 30154785 PMCID: PMC6102393 DOI: 10.3389/fimmu.2018.01818] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/24/2018] [Indexed: 12/11/2022] Open
Abstract
Necroptosis is a pro-inflammatory cell death, which happens in the context of caspase-8 inhibition, allowing activation of the receptor interacting protein kinase 1-receptor interacting protein kinase 3-mixed lineage kinase domain-like (RIPK1-RIPK3-MLKL) axis. Recently, necroptosis has emerged as a key component of resistance against pathogens including infected macrophage by Leishmania infantum, the ethiologic agent of Visceral leishmaniasis (VL). VL is the most severe form of Leishmaniasis, characterized by systemic inflammation and neutropenia. However, the role of neutrophil cell death in VL has not been characterized. Here, we showed that VL patients exhibited increased lactate dehydrogenase levels in the serum, a hallmark of cell death and tissue damage. We investigated the effect of necroptosis in neutrophil infection in vitro. Human neutrophils pretreated with zVAD-fmk (pan-caspase inhibitor) and zIETD-fmk (caspase-8 inhibitor) increased reactive oxygen species (ROS) level in response to Leishmania infection, which is associated with necroptotic cell death. MLKL, an important effector molecule downstream of necroptosis pathway, was also required for Leishmania killing. Moreover, in absence of caspases-8, murine neutrophils displayed loss of membrane integrity, higher levels of ROS, and decreased L. infantum viability. Pharmacological inhibition of RIPK1 or RIPK3 increased parasite survival when caspase-8 was blocked. Electron microscopy assays revealed morphological features associated with necroptotic death in L. infantum infected-neutrophils pretreated with caspase inhibitor, whereas infected cells pretreated with RIPK1 and RIPK3 inhibitors did not show ultra-structural alterations in membrane integrity and presented viable Leishmania within parasitophorous vacuoles. Taken together, these findings suggest that inhibition of caspase-8 contributes to elimination of L. infantum in neutrophils by triggering necroptosis. Thus, targeting necroptosis may represent a new strategy to control Leishmania replication.
Collapse
Affiliation(s)
| | | | | | | | | | - Nivea F Luz
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Graziele Quintela-Carvalho
- Universidade Federal da Bahia, Salvador, Brazil.,Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Instituto Federal de Educação, Ciência e Tecnologia Baiano, Santa Inês, Brazil
| | - Jonilson B Lima
- Centro de Ciências Biológicas e da Saúde, Universidade do Oeste da Bahia, Barreiras, Brazil
| | - Roque P Almeida
- Departamento de Medicina, Universidade Federal de Sergipe, Aracaju, Brazil
| | - Francis K Chan
- Department of Pathology, Immunology and Microbiology Program, University of Massachusetts Medical School, Worcester, MA, United States
| | - Marcelo T Bozza
- Departamento de Imunologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Valeria M Borges
- Universidade Federal da Bahia, Salvador, Brazil.,Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Deboraci B Prates
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Departamento de Biomorfologia, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| |
Collapse
|
25
|
Teixeira CR, Santos CDS, Prates DB, Dos Santos RT, Araújo-Santos T, de Souza-Neto SM, Borges VM, Barral-Netto M, Brodskyn CI. Lutzomyia longipalpis Saliva Drives Interleukin-17-Induced Neutrophil Recruitment Favoring Leishmania infantum Infection. Front Microbiol 2018; 9:881. [PMID: 29867796 PMCID: PMC5953329 DOI: 10.3389/fmicb.2018.00881] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 04/17/2018] [Indexed: 11/22/2022] Open
Abstract
During bloodfeeding, the presence of sand fly saliva in the hemorrhagic pool where Leishmania is also inoculated modulates the development of host immune mechanisms creating a favorable environment for disease progression. To date, information obtained through experimental models suggests that sand fly saliva induces cellular recruitment and modulates production of eicosanoids. However, the effect of sand fly saliva in the different steps of the inflammatory response triggered by Leishmania remains undefined. Here we further investigate if interaction of Lutzomyia longipalpis salivary gland sonicate (SGS) with different host cells present during the initial inflammatory events regulate Leishmania infantum infectivity. Initially, we observed that incubation of human peripheral blood mononuclear cells (PBMC) with Lu. longipalpis SGS in the presence of L. infantum significantly increased IL-10 but did not alter expression of IFN-γ and TNF-α by CD4+ T cells induced by the parasite alone. Interestingly, incubation of PBMC with Lu. longipalpis SGS alone or in the presence of L. infantum resulted in increased IL-17 production. The presence of IL-17 is related to neutrophil recruitment and plays an important role at the site of infection. Here, we also observed increased migration of neutrophil using an in vitro chemotactic assay following incubation with supernatants from PBMC stimulated with L. infantum and Lu. longipalpis SGS. Neutrophil migration was abrogated following neutralization of IL-17 with specific antibodies. Moreover, culture of human neutrophils with L. infantum in the presence of Lu. longipalpis SGS promoted neutrophil apoptosis resulting in increased parasite viability. Neutrophils operate as the first line of defense in the early stages of infection and later interact with different cells, such as macrophages. The crosstalk between neutrophils and macrophages is critical to determine the type of specific immune response that will develop. Here, we observed that co-culture of human macrophages with autologous neutrophils previously infected in the presence of Lu. longipalpis SGS resulted in a higher infection rate, accompanied by increased production of TGF-β and PGE2. Our results provide new insight into the contribution of Lu. longipalpis SGS to L. infantum-induced regulation of important inflammatory events, creating a favorable environment for parasite survival inside different host cells.
Collapse
Affiliation(s)
| | | | - Deboraci B Prates
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Instituto de Ciências da Saúde da Universidade Federal da Bahia, Departamentos de Biomorfologia e Biointeração, Salvador, Brazil
| | | | - Théo Araújo-Santos
- Centro de Ciências Biológicas e Saúde, Universidade Federal do Oeste da Bahia, Barreiras, Brazil
| | | | - Valéria M Borges
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Manoel Barral-Netto
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Faculdade de Medicina da Universidade Federal da Bahia, Departamento de Patologia e Medicina Legal, Salvador, Brazil.,Instituto de Investigação em Imunologia, iii-INCT, São Paulo, Brazil
| | - Cláudia I Brodskyn
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Instituto de Ciências da Saúde da Universidade Federal da Bahia, Departamentos de Biomorfologia e Biointeração, Salvador, Brazil.,Instituto de Investigação em Imunologia, iii-INCT, São Paulo, Brazil
| |
Collapse
|
26
|
Salguero FJ, Garcia-Jimenez WL, Lima I, Seifert K. Histopathological and immunohistochemical characterisation of hepatic granulomas in Leishmania donovani-infected BALB/c mice: a time-course study. Parasit Vectors 2018; 11:73. [PMID: 29386047 PMCID: PMC5793367 DOI: 10.1186/s13071-018-2624-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 01/08/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Visceral leishmaniasis (VL) is a neglected tropical disease (NTD), caused by the intracellular protozoan parasites Leishmania donovani and Leishmania infantum. Symptomatic VL is considered fatal when left untreated. At present, there is no effective vaccine licensed for human use and available chemotherapies have limitations. Understanding the local immune mechanisms required for the control of infection is a key factor for developing effective vaccines and therapeutics. METHODS We have investigated the development of the typical granulomatous lesions in the liver in experimental VL over time, together with the local immune responses. BALB/c mice were infected intravenously with a dose of 2 × 107 L. donovani amastigotes (MHOM/ET/67/HU3) and sacrificed at 15, 35 and 63 days post-infection (dpi). Histopathology and immunohistochemical techniques were used for the detection of Leishmania antigen, selected cell types including B and T lymphocytes, macrophages and neutrophils (CD45R-B220+, CD3+, F4/80+ and Ly-6G+) and iNOS. RESULTS Granulomatous lesions were identified as early as 15 dpi in the livers of all infected animals. Three categories were used to classify liver granulomas (immature, mature and clear). Clear granulomas were exclusively detected from 35 dpi onwards. Kupffer cells (F4/80+) were predominant in immature granulomas, regardless of the dpi. Nonetheless, the highest expression was found 63 dpi. Positive staining for iNOS was mainly observed in the cytoplasm of fused Kupffer cells and the highest expression observed at 35 dpi. T cells (CD3+) and B cells (CD45R-B220+) were predominant in more advanced granuloma stages, probably related to the establishment of acquired immunity. Neutrophils (Ly-6G+) were predominantly observed in mature granulomas with the highest expression at 15 dpi. Neutrophils were lower in numbers compared to other cell types, particularly at later time points. CONCLUSIONS Our results reflect the role of macrophages during the early stage of infection and the establishment of a lymphocytic response to control the infection in more advanced stages.
Collapse
Affiliation(s)
- Francisco J Salguero
- Department of Pathology and Infectious Diseases, School of Veterinary Medicine, University of Surrey, Guildford, UK
| | - Waldo L Garcia-Jimenez
- Department of Pathology and Infectious Diseases, School of Veterinary Medicine, University of Surrey, Guildford, UK
| | - Isadora Lima
- Department of Pathology and Infectious Diseases, School of Veterinary Medicine, University of Surrey, Guildford, UK.,Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz, Salvador, Bahia, Brazil
| | - Karin Seifert
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK.
| |
Collapse
|
27
|
Sabbagh A, Sonon P, Sadissou I, Mendes-Junior CT, Garcia A, Donadi EA, Courtin D. The role of HLA-G in parasitic diseases. HLA 2018; 91:255-270. [PMID: 29368453 DOI: 10.1111/tan.13196] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 12/20/2017] [Indexed: 02/07/2023]
Abstract
Little attention has been devoted to the role of HLA-G gene and molecule on parasitic disorders, and the available studies have focused on malaria, African and American trypanosomiasis, leishmaniosis, toxoplasmosis and echinococcosis. After reporting a brief description regarding the role of the cells of innate and adaptive immune system against parasites, we reviewed the major features of the HLA-G gene and molecule and the role of HLA-G on the major cells of immune system. Increased levels of soluble HLA-G (sHLA-G) have been observed in patients presenting toxoplasmosis and in the active phase of echinococcosis. In addition, increased sHLA-G has also been associated with increased susceptibility to malaria and increased susceptibility to develop human African trypanosomiasis (HAT). In contrast, decreased membrane-bound HLA-G has been reported in placenta of patients infected with Plasmodium falciparum and in heart and colon of patients presenting Chagas disease. The 3' untranslated region of the HLA-G gene has been the main focus of studies on malaria, HAT and Chagas disease, exhibiting distinct patterns of associations. Considering that HLA-G is an immune checkpoint molecule, inhibiting the activity of several cells of the immune system, the excessive neoexpression and the increased sHLA-G levels together with the decreased constitutive tissue expression of membrane-bound HLA-G may be detrimental to the host infected with parasite agents.
Collapse
Affiliation(s)
- A Sabbagh
- UMR 216 MERIT, Institut de Recherche pour le Développement, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France
| | - P Sonon
- Department of Medicine, School of Medicine of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - I Sadissou
- Department of Medicine, School of Medicine of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - C T Mendes-Junior
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - A Garcia
- UMR 216 MERIT, Institut de Recherche pour le Développement, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France.,Centre d'Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l'Enfance (CERPAGE), Faculté des Sciences de la Santé, Cotonou, Bénin
| | - E A Donadi
- Department of Medicine, School of Medicine of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - D Courtin
- UMR 216 MERIT, Institut de Recherche pour le Développement, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France
| |
Collapse
|
28
|
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
|
29
|
Ly6C hi inflammatory monocytes promote susceptibility to Leishmania donovani infection. Sci Rep 2017; 7:14693. [PMID: 29089636 PMCID: PMC5665970 DOI: 10.1038/s41598-017-14935-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/18/2017] [Indexed: 12/24/2022] Open
Abstract
Ly6Chi inflammatory monocytes (iMO) are critical for host defense against toxoplasmosis and malaria but their role in leishmaniasis is unclear. In this study, we report a detrimental role of Ly6Chi iMOs in visceral leishmaniasis (VL) caused by Leishmania donovani. We demonstrate that Ly6Chi iMOs are continuously recruited into the spleen and liver during L. donovani infection and they are preferential targets for the parasite. Using microarray-based gene expression profiling, we show that Ly6Chi iMOs isolated from the infected liver and spleen have distinct phenotypic and activation profiles. Furthermore, we demonstrate that blocking the recruitment of Ly6Chi iMOs into the liver and spleen during L. donovani infection using a CCR2 antagonist reduces the frequency of the pathogenic IFN-γ/IL10 dual producer CD4+ T cells in the spleen and leads to a significant reduction in parasite loads in the liver and spleen. Using STAT1−/− mice we show that STAT1 is critical for mediating the recruitment of Ly6Chi iMOs into organs during L. donovani infection, and adaptive transfer of wild type Ly6Chi iMOs into STAT1−/− recipients renders them susceptible to disease. Our findings reveal an unexpected pathogenic role for Ly6Chi iMOs in promoting parasite survival in VL and open the possibility of targeting this population for host-directed therapy during VL.
Collapse
|
30
|
Scharfstein J, Ramos PIP, Barral-Netto M. G Protein-Coupled Kinin Receptors and Immunity Against Pathogens. Adv Immunol 2017; 136:29-84. [PMID: 28950949 DOI: 10.1016/bs.ai.2017.05.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
For decades, immunologists have considered the complement system as a paradigm of a proteolytic cascade that, acting cooperatively with the immune system, enhances host defense against infectious organisms. In recent years, advances made in thrombosis research disclosed a functional link between activated neutrophils, monocytes, and platelet-driven thrombogenesis. Forging a physical barrier, the fibrin scaffolds generated by synergism between the extrinsic and intrinsic (contact) pathways of coagulation entrap microbes within microvessels, limiting the systemic spread of infection while enhancing the clearance of pathogens by activated leukocytes. Insight from mice models of thrombosis linked fibrin formation via the intrinsic pathway to the autoactivation of factor XII (FXII) by negatively charged "contact" substances, such as platelet-derived polyphosphates and DNA from neutrophil extracellular traps. Following cleavage by FXIIa, activated plasma kallikrein (PK) initiates inflammation by liberating the nonapeptide bradykinin (BK) from an internal domain of high molecular weight kininogen (HK). Acting as a paracrine mediator, BK induces vasodilation and increases microvascular permeability via activation of endothelial B2R, a constitutively expressed subtype of kinin receptor. During infection, neutrophil-driven extravasation of plasma fuels inflammation via extravascular activation of the kallikrein-kinin system (KKS). Whether liberated by plasma-borne PK, tissue kallikrein, and/or microbial-derived proteases, the short-lived kinins activate immature dendritic cells via B2R, thus linking the infection-associated innate immunity/inflammation to the adaptive arm of immunity. As inflammation persists, a GPI-linked carboxypeptidase M removes the C-terminal arginine from the primary kinin, converting the B2R agonist into a high-affinity ligand for B1R, a GPCR subtype that is transcriptionally upregulated in injured/inflamed tissues. As reviewed here, lessons taken from studies of kinin receptor function in experimental infections have shed light on the complex proteolytic circuits that, acting at the endothelial interface, reciprocally couple immunity to the proinflammatory KKS.
Collapse
Affiliation(s)
- Julio Scharfstein
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Center of Health Sciences (CCS), Cidade Universitária, Rio de Janeiro, Brazil.
| | - Pablo I P Ramos
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
| | | |
Collapse
|
31
|
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
|
32
|
Immunization with Leishmania donovani protein disulfide isomerase DNA construct induces Th1 and Th17 dependent immune response and protection against experimental visceral leishmaniasis in Balb/c mice. Mol Immunol 2017; 82:104-113. [DOI: 10.1016/j.molimm.2016.12.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 12/20/2016] [Accepted: 12/23/2016] [Indexed: 11/20/2022]
|
33
|
Gamma Interferon-Regulated Chemokines in Leishmania donovani Infection in the Liver. Infect Immun 2016; 85:IAI.00824-16. [PMID: 27795366 DOI: 10.1128/iai.00824-16] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 10/16/2016] [Indexed: 12/25/2022] Open
Abstract
In the livers of C57BL/6 mice, gamma interferon (IFN-γ) controls intracellular Leishmania donovani infection and the efficacy of antimony (Sb) chemotherapy. Since both responses usually correlate with granulomatous inflammation, we tested six prominently expressed, IFN-γ-regulated chemokines-CXCL9, CXCL10, CXCL13, CXCL16, CCL2, and CCL5-for their roles in (i) mononuclear cell recruitment and granuloma assembly and maturation, (ii) initial control of infection and self-cure, and (iii) responsiveness to Sb treatment. Together, the results for the L. donovani-infected livers of chemokine-deficient mice (CXCR6-/- mice were used as CXCL16-deficient surrogates) indicated that individual IFN-γ-induced chemokines have diverse affects and (i) may be entirely dispensable (CXCL13, CXCL16), (ii) may promote (CXCL10, CCL2, CCL5) or downregulate (CXCL9) initial granuloma assembly, (iii) may enhance (CCL2, CCL5) or hinder (CXCL10) early parasite control, (iv) may promote granuloma maturation (CCL2, CCL5), (v) may exert a granuloma-independent action that enables self-cure (CCL5), and (vi) may have no role in responsiveness to chemotherapy. Despite the near absence of tissue inflammation in early-stage infection, parasite replication could be controlled (in CXCL10-/- mice) and Sb was fully active (in CXCL10-/-, CCL2-/-, and CCL5-/- mice). These results characterize chemokine action in the response to L. donovani and also reemphasize that (i) recruited mononuclear cells and granulomas are not required to control infection or respond to Sb chemotherapy, (ii) granuloma assembly, control of infection, and Sb's efficacy are not invariably linked expressions of the same T cell-dependent, cytokine-mediated antileishmanial mechanism, and (iii) granulomas are not necessarily hallmarks of protective antileishmanial immunity.
Collapse
|
34
|
Davis RE, Sharma S, Conceição J, Carneiro P, Novais F, Scott P, Sundar S, Bacellar O, Carvalho EM, Wilson ME. Phenotypic and functional characteristics of HLA-DR + neutrophils in Brazilians with cutaneous leishmaniasis. J Leukoc Biol 2016; 101:739-749. [PMID: 28076241 DOI: 10.1189/jlb.4a0915-442rr] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Revised: 09/08/2016] [Accepted: 09/25/2016] [Indexed: 11/24/2022] Open
Abstract
The protozoan Leishmania braziliensis causes cutaneous leishmaniasis (CL) in endemic regions. In murine models, neutrophils (PMNs) are recruited to the site of infection soon after parasite inoculation. However, the roles of neutrophils during chronic infection and in human disease remain undefined. We hypothesized that neutrophils help maintain a systemic inflammatory state in subjects with CL. Lesion biopsies from all patients with CL tested contained neutrophils expressing HLA-DR, a molecule thought to be restricted to professional antigen-presenting cells. Although CL is a localized disease, a subset of patients with CL also had circulating neutrophils expressing HLA-DR and the costimulatory molecules CD80, CD86, and CD40. PMNs isolated from a low-density leukocyte blood fraction (LD-PMNs) contained a higher percentage of HLA-DR+ PMNs than did normal-density PMNs. In vitro coculture experiments suggested LD-PMNs do not suppress T cell responses, differentiating them from MDSCs. Flow-sorted HLA-DR+ PMNs morphologically resembled conventional PMNs, and they exhibited functional properties of PMNs. Compared with conventional PMNs, HLA-DR+ PMNs showed increased activation, degranulation, DHR123 oxidation, and phagocytic capacity. A few HLA-DR+ PMNs were observed in healthy subjects, and that proportion could be increased by incubation in either inflammatory cytokines or in plasma from a patient with CL. This was accompanied by an increase in PMN hladrb1 mRNA, suggesting a possible connection between neutrophil "priming" and up-regulation of HLA-DR. These data suggest that PMNs that are primed for activation and that also express surface markers of antigen-presenting cells emerge in the circulation and infected tissue lesions of patients with CL.
Collapse
Affiliation(s)
- Richard E Davis
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Smriti Sharma
- Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Jacilara Conceição
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil.,Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais, INCT-DT (CNPq/MCT), Salvador, Bahia, Brazil
| | - Pedro Carneiro
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil.,Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais, INCT-DT (CNPq/MCT), Salvador, Bahia, Brazil
| | - Fernanda Novais
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Shyam Sundar
- Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Olivia Bacellar
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil.,Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais, INCT-DT (CNPq/MCT), Salvador, Bahia, Brazil
| | - Edgar M Carvalho
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil.,Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais, INCT-DT (CNPq/MCT), Salvador, Bahia, Brazil.,Fundação Gonçalo Muniz, Fiocruz-Bahia, Salvador, Bahia Brazil
| | - Mary E Wilson
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA; .,Departments of Internal Medicine and Microbiology, University of Iowa, Iowa City, Iowa, USA; and.,Research Service, Iowa City Veterans' Affairs Medical Center, Iowa City, Iowa, USA
| |
Collapse
|
35
|
de Mendonça LZ, Resende LA, Lanna MF, Aguiar-Soares RDDO, Roatt BM, Castro RADOE, Batista MA, Silveira-Lemos D, Gomes JDAS, Fujiwara RT, Rezende SA, Martins-Filho OA, Corrêa-Oliveira R, Dutra WO, Reis AB, Giunchetti RC. Multicomponent LBSap vaccine displays immunological and parasitological profiles similar to those of Leish-Tec® and Leishmune® vaccines against visceral leishmaniasis. Parasit Vectors 2016; 9:472. [PMID: 27577735 PMCID: PMC5006379 DOI: 10.1186/s13071-016-1752-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 08/12/2016] [Indexed: 11/10/2022] Open
Abstract
Background In past years, many researchers have sought canine visceral leishmaniasis (CVL) prevention through the characterization of Leishmania antigens as vaccine candidates. Despite these efforts, there is still no efficient vaccine for CVL control. Methods In the present study, we performed a pre-clinical vaccine trial using BALB/c mice to compare the effects of the multicomponent LBSap vaccine with those of Leish-Tec® and Leishmune®. Blood was collected to determine the frequency of peripheral blood cells and to evaluate hematologic and immunophenotypic parameters. Liver and spleen samples were collected for parasitological quantification, and spleen samples were used to access the cytokine profile. Results When measuring total IgG and IgG1 anti-Leishmania levels after the third vaccination and L. infantum challenge, it was evident that all vaccines were able to induce humoral immune response. Regarding the innate immune response, increased levels of NK CD3-CD49+ cells were the hallmark of all vaccinated groups, whereas only the Leish-Tec® group displayed a high frequency of CD14+ monocytes after L. infantum challenge. Moreover, CD3+CD4+ T cells were the main circulating lymphocytes induced after L. infantum challenge with all evaluated vaccines. Importantly, after L. infantum challenge, splenocytes from the Leishmune® vaccine produced high levels of IL-2, whereas a prominent type 1 immune response was the hallmark of the LBSap vaccine, which presented high levels of IL-2, IL-6, TNF-α, and IFN-γ. The efficacy analysis using real-time polymerase chain reaction demonstrated a reduction in the parasitism in the spleen (Leishmune®: 64 %; LBSap: 42 %; and Leish-Tec®: 36 %) and liver (Leishmune®: 71 %; LBSap: 62 %; and Leish-Tec®: 48 %). Conclusions The dataset led to the conclusion that the LBSap vaccination was able to induce immune and efficacy profiles comparable with those of commercial vaccines, thus demonstrating its potential as a promising vaccine candidate for visceral leishmaniasis control. Electronic supplementary material The online version of this article (doi:10.1186/s13071-016-1752-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ludmila Zanandreis de Mendonça
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Lucilene Aparecida Resende
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas/NUPEB, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Mariana Ferreira Lanna
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas/NUPEB, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Rodrigo Dian de Oliveira Aguiar-Soares
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas/NUPEB, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Bruno Mendes Roatt
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas/NUPEB, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Renata Alves de Oliveira E Castro
- Laboratório de Pesquisas Clínicas, Programa de Pós-Graduação de Ciências Farmacêuticas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Maurício Azevedo Batista
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Denise Silveira-Lemos
- Laboratório de Biomarcadores de Diagnóstico e Monitoração, Centro de Pesquisa René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Juliana de Assis Silva Gomes
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Simone Aparecida Rezende
- Laboratório de Pesquisas Clínicas, Programa de Pós-Graduação de Ciências Farmacêuticas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Olindo Assis Martins-Filho
- Laboratório de Biomarcadores de Diagnóstico e Monitoração, Centro de Pesquisa René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Rodrigo Corrêa-Oliveira
- Laboratório de Imunologia Celular e Molecular, Centro de Pesquisa René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Walderez Ornelas Dutra
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Alexandre Barbosa Reis
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas/NUPEB, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Rodolfo Cordeiro Giunchetti
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
36
|
Interleukin-27 (IL-27) Mediates Susceptibility to Visceral Leishmaniasis by Suppressing the IL-17-Neutrophil Response. Infect Immun 2016; 84:2289-2298. [PMID: 27245409 DOI: 10.1128/iai.00283-16] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/21/2016] [Indexed: 01/01/2023] Open
Abstract
The relationship established between Leishmania infantum and the vertebrate host can lead to a self-healing infection or to the manifestation of visceral leishmaniasis, a chronic systemic infection associated with high rates of mortality. We hypothesized that regulatory cytokines, such as interleukin-27 (IL-27), play a role in susceptibility to L. infantum infection. IL-27 is a heterodimeric cytokine composed of IL-27p28 and EBi3 subunits which, when combined, bind to IL-27R, leading to STAT-1 and -3 activation, playing a role in the regulation of the immune response. We observed in this work that IL-27 regulates the Th1/Th17 profiles in a mouse model of visceral leishmaniasis (VL) caused by L. infantum We showed here that the pathogen recognition by endosomal Toll-like receptors triggers a type I interferon (IFN) response, which acts through the type I IFN receptor and interferon regulatory factor 1 to induce IL-27 production by macrophages. Furthermore, IL-27 plays a major regulatory role in vivo, because Ebi3(-/-) mice can efficiently control parasite replication despite reduced levels of IFN-γ compared to wild-type mice. On the other hand, the absence of Ebi3 leads to exacerbated IL-17A production in the infected organs as well as in a coculture system, suggesting a direct regulatory action of IL-27 during L. infantum infection. As a consequence of exacerbated IL-17A in Ebi3(-/-) mice, a greater neutrophil influx was observed in the target organs, playing a role in parasite control. Thus, this work unveiled the molecular steps of IL-27 production after L. infantum infection and demonstrated its regulatory role in the IL-17A-neutrophil axis.
Collapse
|
37
|
Banerjee A, Bhattacharya P, Joshi AB, Ismail N, Dey R, Nakhasi HL. Role of pro-inflammatory cytokine IL-17 in Leishmania pathogenesis and in protective immunity by Leishmania vaccines. Cell Immunol 2016; 309:37-41. [PMID: 27444130 DOI: 10.1016/j.cellimm.2016.07.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 06/28/2016] [Accepted: 07/06/2016] [Indexed: 02/01/2023]
Abstract
The clinical outcome of Leishmania pathogenesis ranges from active skin lesions to fatal visceral dissemination and severely impaired T cell immunity. It is well established that a strong Th1 immune response is protective against cutaneous forms of the disease, however a mixed Th1/Th2 response is most commonly observed against visceral infections as evident from previous studies. Aside from Th1/Th2 cytokines, the pro-inflammatory IL-17 cytokine family plays an important role in the clearance of intracellular pathogens. In Leishmania induced skin lesions, IL-17 produced by Th17 cells is shown to exacerbate the disease, suggesting a role in pathogenesis. However, a protective role for IL-17 is indicated by the expansion of IL-17 producing cells in vaccine-induced immunity. In human visceral leishmaniasis (VL) it has been demonstrated that IL-17 and IL-22 are associated with protection against re-exposure to Leishmania, which further suggests the involvement of IL-17 in vaccine induced protective immunity. Although there is no vaccine against any form of leishmaniasis, the development of genetically modified live attenuated parasites as vaccine candidates prove to be promising, as they successfully induce a robust protective immune response in various animal models. However, the role of IL-17 producing cells and Th17 cells in response to these vaccine candidates remains unexplored. In this article, we review the role of IL-17 in Leishmania pathogenesis and the potential impact on vaccine induced immunity, with a special focus on live attenuated Leishmania parasites.
Collapse
Affiliation(s)
- Antara Banerjee
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - 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, USA
| | - Amritanshu B Joshi
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Nevien Ismail
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - 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, USA.
| | - 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, USA.
| |
Collapse
|
38
|
Srivastava S, Shankar P, Mishra J, Singh S. Possibilities and challenges for developing a successful vaccine for leishmaniasis. Parasit Vectors 2016; 9:277. [PMID: 27175732 PMCID: PMC4866332 DOI: 10.1186/s13071-016-1553-y] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 04/28/2016] [Indexed: 02/07/2023] Open
Abstract
Leishmaniasis is a vector-borne disease caused by different species of protozoan parasites of the genus Leishmania. It is a major health problem yet neglected tropical diseases, with approximately 350 million people worldwide at risk and more than 1.5 million infections occurring each year. Leishmaniasis has different clinical manifestations, including visceral (VL or kala-azar), cutaneous (CL), mucocutaneous (MCL), diffuse cutaneous (DCL) and post kala-azar dermal leishmaniasis (PKDL). Currently, the only mean to treat and control leishmaniasis is by rational medications and vector control. However, the number of available drugs is limited and even these are either exorbitantly priced, have toxic side effects or prove ineffective due to the emergence of resistant strains. On the other hand, the vector control methods are not so efficient. Therefore, there is an urgent need for developing a safe, effective, and affordable vaccine for the prevention of leishmaniasis. Although in recent years a large body of researchers has concentrated their efforts on this issue, yet only three vaccine candidates have gone for clinical trial, until date. These are: (i) killed vaccine in Brazil for human immunotherapy; (ii) live attenuated vaccine for humans in Uzbekistan; and (iii) second-generation vaccine for dog prophylaxis in Brazil. Nevertheless, there are at least half a dozen vaccine candidates in the pipeline. One can expect that, in the near future, the understanding of the whole genome of Leishmania spp. will expand the vaccine discovery and strategies that may provide novel vaccines. The present review focuses on the development and the status of various vaccines and potential vaccine candidates against leishmaniasis.
Collapse
Affiliation(s)
- Saumya Srivastava
- Division of Clinical Microbiology and Molecular Medicine, Department of Laboratory Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Prem Shankar
- Division of Clinical Microbiology and Molecular Medicine, Department of Laboratory Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Jyotsna Mishra
- Division of Clinical Microbiology and Molecular Medicine, Department of Laboratory Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Sarman Singh
- Division of Clinical Microbiology and Molecular Medicine, Department of Laboratory Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India.
| |
Collapse
|
39
|
Davis RE, Thalhofer CJ, Wilson ME. Infection and Activation of Human Neutrophils with Fluorescent Leishmania infantum. ACTA ACUST UNITED AC 2016; 5. [PMID: 30381805 DOI: 10.4172/2329-9541.1000146] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neutrophils (PMNs) are recruited in high numbers to sites of host infection by the protozoan parasites of the genus Leishmania. Although PMNs are capable of phagocytizing Leishmania parasites and are potent producers of anti-microbial compounds including reactive oxygen species (ROS), they are unable to control the establishment of infection. Prior studies document production of ROS in isolated PMNs incubated with Leishmania under conditions allowing phagocytosis, but without a measure of single cells' responses it cannot be discerned whether PMN activation and ROS production is suppressed or ineffective in the cells that internalize the parasite. To address these interactions, we engineered a strain of fluorescent, mCherry-expressing Leishmania infantum (mCherry-Li). By infecting isolated human PMNs in vitro with mCherry-Li, we observed ready association of the parasites with PMNs in a time- and dose-dependent fashion. We also examined production of PMN ROS (using the fluorescent compound DHR123) and PMN activation (as evidence by loss of surface CD62L expression). Whereas many Li-associated (mCherry+) PMNs responded to parasite interactions and uptake with ROS production and/or activation, a proportion exhibited neither response. Furthermore, a large proportion of mCherry - "bystander" PMNs displayed both ROS production and activation. The heterogeneous response of PMNs to Leishmania exposure leads us to hypothesize, first, that some PMNs exhibit decreased activation upon phagocytosis of Leishmania, and could support their maintenance. Second, responses of bystander PMNs may contribute to a local inflammatory environment that is ineffective at parasite clearance.
Collapse
Affiliation(s)
- R E Davis
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA, USA
| | | | - M E Wilson
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA, USA.,Departments of Internal Medicine and Microbiology, University of Iowa, Iowa City, IA, USA.,Veterans Affairs Medical Center, Iowa City, IA, USA
| |
Collapse
|
40
|
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]
|
41
|
de Freitas EO, Leoratti FMDS, Freire-de-Lima CG, Morrot A, Feijó DF. The Contribution of Immune Evasive Mechanisms to Parasite Persistence in Visceral Leishmaniasis. Front Immunol 2016; 7:153. [PMID: 27148272 PMCID: PMC4840207 DOI: 10.3389/fimmu.2016.00153] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/08/2016] [Indexed: 01/06/2023] Open
Abstract
Leishmania is a genus of protozoan parasites that give rise to a range of diseases called Leishmaniasis that affects annually an estimated 1.3 million people from 88 countries. Leishmania donovani and Leishmania (L.) infantum chagasi are responsible to cause the visceral leishmaniasis. The parasite can use assorted strategies to interfere with the host homeostasis to establish persistent infections that without treatment can be lethal. In this review, we highlight the mechanisms involved in the parasite subversion of the host protective immune response and how alterations of host tissue physiology and vascular remodeling during VL could affect the organ-specific immunity against Leishmania parasites.
Collapse
Affiliation(s)
| | | | | | - Alexandre Morrot
- Laboratorio de Biologia do Sistema Imune, Departmento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro , Rio de Janeiro , Brazil
| | - Daniel Ferreira Feijó
- Laboratório Integrado de Microbiologia e Imunoregulação, Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ) , Salvador , Brazil
| |
Collapse
|
42
|
Maran N, Gomes PS, Freire-de-Lima L, Freitas EO, Freire-de-Lima CG, Morrot A. Host resistance to visceral leishmaniasis: prevalence and prevention. Expert Rev Anti Infect Ther 2016; 14:435-42. [DOI: 10.1586/14787210.2016.1160779] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
43
|
Kaye PM, Beattie L. Lessons from other diseases: granulomatous inflammation in leishmaniasis. Semin Immunopathol 2015; 38:249-60. [PMID: 26678994 PMCID: PMC4779128 DOI: 10.1007/s00281-015-0548-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 11/02/2015] [Indexed: 11/16/2022]
Abstract
The Leishmania granuloma shares some, though not all, properties with that formed following mycobacterial infection. As a simplified, noncaseating granuloma composed of relatively few and largely mononuclear cell populations, it provides a tractable model system to investigate intra-granuloma cellular dynamics, immune regulation, and antimicrobial resistance. Here, the occurrence of granulomatous pathology across the spectrum of leishmaniasis, in humans and animal reservoir hosts, is first described. However, this review focuses on the process of hepatic granuloma formation as studied in rodent models of visceral leishmaniasis, starting from the initial infection of Kupffer cells to the involution of the granuloma after pathogen clearance. It describes how the application of intravital imaging and the use of computational modeling have changed some of our thoughts on granuloma function, and illustrates how host-directed therapies have been used to manipulate granuloma form and function for therapeutic benefit. Where appropriate, lessons that may be equally applicable across the spectrum of granulomatous diseases are highlighted.
Collapse
Affiliation(s)
- Paul M Kaye
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, Heslington, York, YO10 5DD, UK.
| | - Lynette Beattie
- QIMR Berghofer Medical Research Institute, 300 Herston Rd, Herston, Queensland, Australia, 4006
| |
Collapse
|
44
|
Terrazas C, Varikuti S, Kimble J, Moretti E, Boyaka PN, Satoskar AR. IL-17A promotes susceptibility during experimental visceral leishmaniasis caused by Leishmania donovani. FASEB J 2015; 30:1135-43. [PMID: 26581600 DOI: 10.1096/fj.15-277202] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 11/09/2015] [Indexed: 12/20/2022]
Abstract
Leishmania donovani is an intracellular parasite that infects professional phagocytes and causes visceral leishmaniasis (VL). The immune response during VL has been extensively studied in the context of T-helper (Th)1 and Th2 responses. Immunity against this parasite is dependent on IFN-γ production and subsequent macrophage activation, and the Th2 response promotes granuloma formation. The cytokine IL-17A is associated with neutrophilic inflammation. Depletion of neutrophils during experimental VL results in enhanced parasitic loads. Furthermore, although patients resistant to VL showed enhanced levels of IL-17A in circulation, little is known about the role of IL-17A during VL infection. Here, we used IL-17A-deficient mice and IL-17A reporter mice to address the role of IL-17A during VL. IL-17A(-/-) mice were highly resistant to VL infection, showing decreased parasites in the liver and spleen. This unexpected phenotype was associated with enhanced IFN-γ production by T cells and decreased accumulation of neutrophils and monocytes, resulting in reduced number of granulomas. We also found γδ T and Th17 cells as the main IL-17A(+) cells during VL infection. Our data reveal an unexpected role of IL-17A rendering susceptibility against L. donovani by regulating the IFN-γ response and promoting detrimental inflammation.
Collapse
Affiliation(s)
- Cesar Terrazas
- *Department of Pathology, Department of Veterinary Biosciences, and Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Sanjay Varikuti
- *Department of Pathology, Department of Veterinary Biosciences, and Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Jennifer Kimble
- *Department of Pathology, Department of Veterinary Biosciences, and Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Ellen Moretti
- *Department of Pathology, Department of Veterinary Biosciences, and Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Prosper N Boyaka
- *Department of Pathology, Department of Veterinary Biosciences, and Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Abhay R Satoskar
- *Department of Pathology, Department of Veterinary Biosciences, and Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
45
|
Toll-like receptor 9 signaling in dendritic cells regulates neutrophil recruitment to inflammatory foci following Leishmania infantum infection. Infect Immun 2015; 83:4604-16. [PMID: 26371124 DOI: 10.1128/iai.00975-15] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 09/06/2015] [Indexed: 11/20/2022] Open
Abstract
Leishmania infantum is a protozoan parasite that causes visceral leishmaniasis (VL). This infection triggers dendritic cell (DC) activation through the recognition of microbial products by Toll-like receptors (TLRs). Among the TLRs, TLR9 is required for DC activation by different Leishmania species. We demonstrated that TLR9 is upregulated in vitro and in vivo during infection. We show that C57BL/6 mice deficient in TLR9 expression (TLR9(-/-) mice) are more susceptible to infection and display higher parasite numbers in the spleen and liver. The increased susceptibility of TLR9(-/-) mice was due to the impaired recruitment of neutrophils to the infection foci associated with reduced levels of neutrophil chemoattractants released by DCs in the target organs. Moreover, both Th1 and Th17 cells were also committed in TLR9(-/-) mice. TLR9-dependent neutrophil recruitment is mediated via the MyD88 signaling pathway but is TIR domain-containing adapter-inducing interferon beta (TRIF) independent. Furthermore, L. infantum failed to activate both plasmacytoid and myeloid DCs from TLR9(-/-) mice, which presented reduced surface costimulatory molecule expression and chemokine release. Interestingly, neutrophil chemotaxis was affected both in vitro and in vivo when DCs were derived from TLR9(-/-) mice. Our results suggest that TLR9 plays a critical role in neutrophil recruitment during the protective response against L. infantum infection that could be associated with DC activation.
Collapse
|
46
|
Carlsen ED, Liang Y, Shelite TR, Walker DH, Melby PC, Soong L. Permissive and protective roles for neutrophils in leishmaniasis. Clin Exp Immunol 2015; 182:109-18. [PMID: 26126690 DOI: 10.1111/cei.12674] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2015] [Indexed: 12/11/2022] Open
Abstract
Leishmania parasites are the causative agents of leishmaniasis, a neglected tropical disease that causes substantial morbidity and considerable mortality in many developing areas of the world. Recent estimates suggest that roughly 10 million people suffer from cutaneous leishmaniasis (CL), and approximately 76,000 are afflicted with visceral leishmaniasis (VL), which is universally fatal without treatment. Efforts to develop therapeutics and vaccines have been greatly hampered by an incomplete understanding of the parasite's biology and a lack of clear protective correlates that must be met in order to achieve immunity. Although parasites grow and divide preferentially in macrophages, a number of other cell types interact with and internalize Leishmania parasites, including monocytes, dendritic cells and neutrophils. Neutrophils appear to be especially important shortly after parasites are introduced into the skin, and may serve a dual protective and permissive role during the establishment of infection. Curiously, neutrophil recruitment to the site of infection appears to continue into the chronic phase of disease, which may persist for many years. The immunological impact of these cells during chronic leishmaniasis is unclear at this time. In this review we discuss the ways in which neutrophils have been observed to prevent and promote the establishment of infection, examine the role of anti-neutrophil antibodies in mouse models of leishmaniasis and consider recent findings that neutrophils may play a previously unrecognized role in influencing chronic parasite persistence.
Collapse
Affiliation(s)
- E D Carlsen
- Department of Internal Medicine, Division of Infectious Diseases, MD-PhD Combined Degree Program.,Department of Microbiology and Immunology
| | - Y Liang
- Department of Microbiology and Immunology
| | | | | | - P C Melby
- Department of Microbiology and Immunology.,Department of Pathology.,Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - L Soong
- Department of Microbiology and Immunology.,Department of Pathology
| |
Collapse
|
47
|
New insights into neutrophil and Leishmania infantum in vitro immune interactions. Comp Immunol Microbiol Infect Dis 2015; 40:19-29. [DOI: 10.1016/j.cimid.2015.03.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 02/24/2015] [Accepted: 03/20/2015] [Indexed: 01/09/2023]
|
48
|
Khadem F, Uzonna JE. Immunity to visceral leishmaniasis: implications for immunotherapy. Future Microbiol 2015; 9:901-15. [PMID: 25156379 DOI: 10.2217/fmb.14.43] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Visceral leishmaniasis, caused by Leishmania donovani, L. infantum (syn. Leishmania chagasi), is a globally widespread disease with a burden of about 400,000 new infections reported annually. It is the most dangerous form of human leishmaniasis in terms of mortality and morbidity and is spreading to several nonendemic areas because of migration, global traveling and military conflicts. The emergence of Leishmania-HIV co-infection and increased prevalence of drug-resistant strains have worsened the impact of the disease. The traditional low-cost drugs are often toxic with several adverse effects, highlighting the need for development of new therapeutic and prophylactic strategies. Therefore, a detailed understanding of mechanisms of protective immunity is extremely important in order to develop new therapeutics in the form of vaccines or immunotherapies. This review gives an overview of visceral leishmaniasis, with particular emphasis on the innate and adaptive immune responses, vaccine and vaccination strategies and their potentials for immunotherapy against the disease.
Collapse
Affiliation(s)
- Forough Khadem
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | | |
Collapse
|
49
|
Falcão SAC, Weinkopff T, Hurrell BP, Celes FS, Curvelo RP, Prates DB, Barral A, Borges VM, Tacchini-Cottier F, de Oliveira CI. Exposure to Leishmania braziliensis triggers neutrophil activation and apoptosis. PLoS Negl Trop Dis 2015; 9:e0003601. [PMID: 25756874 PMCID: PMC4354905 DOI: 10.1371/journal.pntd.0003601] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 02/06/2015] [Indexed: 11/21/2022] Open
Abstract
Background Neutrophils are the first line of defense against invading pathogens and are rapidly recruited to the sites of Leishmania inoculation. During Leishmania braziliensis infection, depletion of inflammatory cells significantly increases the parasite load whereas co-inoculation of neutrophils plus L. braziliensis had an opposite effect. Moreover, the co-culture of infected macrophages and neutrophils also induced parasite killing leading us to ask how neutrophils alone respond to an L. braziliensis exposure. Herein we focused on understanding the interaction between neutrophils and L. braziliensis, exploring cell activation and apoptotic fate. Methods and Findings Inoculation of serum-opsonized L. braziliensis promastigotes in mice induced neutrophil accumulation in vivo, peaking at 24 h. In vitro, exposure of thyoglycollate-elicited inflammatory or bone marrow neutrophils to L. braziliensis modulated the expression of surface molecules such as CD18 and CD62L, and induced the oxidative burst. Using mCherry-expressing L. braziliensis, we determined that such effects were mainly observed in infected and not in bystander cells. Neutrophil activation following contact with L. braziliensis was also confirmed by the release of TNF-α and neutrophil elastase. Lastly, neutrophils infected with L. braziliensis but not with L. major displayed markers of early apoptosis. Conclusions We show that L. braziliensis induces neutrophil recruitment in vivo and that neutrophils exposed to the parasite in vitro respond through activation and release of inflammatory mediators. This outcome may impact on parasite elimination, particularly at the early stages of infection. Leishmania is the parasite responsible for the disease leishmaniasis, present in all continents. Leishmania parasites are spread through infected sand-flies and, during transmission into the vertebrate host, neutrophils are among the first cells to arrive at the infection site. Since neutrophils are key players at the frontline of defense against invading organisms, we investigated their response to Leishmania braziliensis. Importantly, L. braziliensis causes both Cutaneous and Mucocutaneous Leishmaniasis, two clinical manifestations characterized by their chronic development and by the presence of skin lesions with tissue destruction. Upon inoculation of mice with L. braziliensis, neutrophils rapidly arrive at the site of infection. We then observed that culture of mouse neutrophils with L. braziliensis induced the expression of adhesion molecules, production of Reactive Oxygen Species and secretion of elastase and TNF-α, two important inflammatory mediators. Also, infection with L. braziliensis induced neutrophil apoptosis, a cell death mechanism key for regulating inflammation. Our results show that neutrophils respond to presence of the L. braziliensis parasites by becoming activated and undergoing apoptosis. We suggest that this outcome modifies the local environment at the site of parasite inoculation and thus contributes with parasite killing in the infected host.
Collapse
Affiliation(s)
| | - Tiffany Weinkopff
- Department of Biochemistry, and WHO-Immunology Research and Training Center, University of Lausanne, Epalinges, Switzerland
| | - Benjamin P. Hurrell
- Department of Biochemistry, and WHO-Immunology Research and Training Center, University of Lausanne, Epalinges, Switzerland
| | - Fabiana S. Celes
- Centro de Pesquisas Gonçalo Moniz, FIOCRUZ, Salvador, Bahia, Brazil
| | | | - Deboraci B. Prates
- Departamento de Biomorfologia, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Aldina Barral
- Centro de Pesquisas Gonçalo Moniz, FIOCRUZ, Salvador, Bahia, Brazil
- Instituto de Investigação em Imunologia, Salvador, Bahia, Brazil
| | - Valeria M. Borges
- Centro de Pesquisas Gonçalo Moniz, FIOCRUZ, Salvador, Bahia, Brazil
- Instituto de Investigação em Imunologia, Salvador, Bahia, Brazil
| | - Fabienne Tacchini-Cottier
- Department of Biochemistry, and WHO-Immunology Research and Training Center, University of Lausanne, Epalinges, Switzerland
| | - Camila I. de Oliveira
- Centro de Pesquisas Gonçalo Moniz, FIOCRUZ, Salvador, Bahia, Brazil
- Instituto de Investigação em Imunologia, Salvador, Bahia, Brazil
- * E-mail:
| |
Collapse
|
50
|
Stein J, Maxeiner JH, Montermann E, Höhn Y, Raker V, Taube C, Sudowe S, Reske-Kunz AB. Non-eosinophilic airway hyper-reactivity in mice, induced by IFN-γ producing CD4(+) and CD8(+) lung T cells, is responsive to steroid treatment. Scand J Immunol 2015; 80:327-38. [PMID: 25124713 DOI: 10.1111/sji.12217] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 08/06/2014] [Indexed: 01/22/2023]
Abstract
Non-eosinophilic asthma is characterized by infiltration of neutrophils into the lung and variable responsiveness to glucocorticoids. The pathophysiological mechanisms have not been characterized in detail. Here, we present an experimental asthma model in mice associated with non-eosinophilic airway inflammation and airway hyper-responsiveness (AHR). For this, BALB/c mice were sensitized by biolistic DNA immunization with a plasmid encoding the model antigen β-galactosidase (pFascin-βGal mice). For comparison, eosinophilic airway inflammation was induced by subcutaneous injection of βGal protein (βGal mice). Intranasal challenge of mice in both groups induced AHR to a comparable extent as well as recruitment of inflammatory cells into the airways. In contrast to βGal mice, which exhibited extensive eosinophilic infiltration in the lung, goblet cell hyperplasia and polarization of CD4(+) T cells into Th2 and Th17 cells, pFascin-βGal mice showed considerable neutrophilia, but no goblet cell hyperplasia and a predominance of Th1 and Tc1 cells in the airways. Depletion studies in pFascin-βGal mice revealed that CD4(+) and CD8(+) cells cooperated to induce maximum inflammation, but that neutrophilic infiltration was not a prerequisite for AHR induction. Treatment of pFascin-βGal mice with dexamethasone before intranasal challenge did not affect neutrophilic infiltration, but significantly reduced AHR, infiltration of monocytes and lymphocytes as well as content of IFN-γ in the bronchoalveolar fluid. Our results suggest that non-eosinophilic asthma associated predominantly with Th1/Tc1 cells is susceptible to glucocorticoid treatment. pFascin-βGal mice might represent a mouse model to study pathophysiological mechanisms proceeding in the subgroup of asthmatics with non-eosinophilic asthma that respond to inhaled steroids.
Collapse
Affiliation(s)
- J Stein
- Clinical Research Unit Allergology, Department of Dermatology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | | | | | | | | | | | | | | |
Collapse
|