1
|
Wojtkowiak-Giera A, Kosik-Bogacka D, Łanocha-Arendarczyk N, Kolasa A, Kot K, Solarczyk P, Derda M. Toll-like receptors and inflammatory cytokines in the skin of Acanthamoeba spp. infected immunocompetent and immunosuppressed mice. Exp Parasitol 2025; 273:108944. [PMID: 40274043 DOI: 10.1016/j.exppara.2025.108944] [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: 10/16/2024] [Revised: 02/02/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Acanthamoeba spp. can cause opportunistic infections, such as cutaneous acanthamoebiasis (CA). Little is known about the role of TLRs and cytokines in the host skin during Acanthamoeba spp. infections. The study aimed to examine the gene and protein expression of TLR3, TLR7, IFN-γ, and IL-23 in the skin of mice experimentally infected with a clinical strain of Acanthamoeba spp. male BALB/c mice were assigned to four groups: group I (control group I) - with normal immunity (C, n = 5); group II (control group II) - with reduced immunity induced by methylprednisolone sodium succinate (MPS; CS, n = 5); group III - amoeba-infected hosts with normal immunity (A, n = 12); and group IV- amoeba-infected hosts with reduced immunity induced by MPS (AS, n = 12). The skin sections (2 cm × 2 cm) were collected from the animals at 8, 16, and 24 days post-infection (dpi). TLR3, TLR7, IFN-γ, and IL-23 gene and protein expressions were analyzed by quantitative real-time PCR and immunohistochemical staining. In the immunocompetent hosts, we noted higher expressions of TLR3 and IL-23 at all-time points, except 8th dpi when IL-23 gene expression was downregulated compared to the control group. The mRNA expressions of TLR7 and IFN-γ were higher at 16 and 24 dpi in the skin of immunocompetent Acanthamoeba spp.-infected hosts than in the uninfected mice. In the course of acanthamoebiasis in the mice with reduced immunity, we found significant upregulation of TLR3, IL-23, and TLR7 gene expressions only at the beginning of infection compared to the control group. A similar relationship was observed for IFN-γ at 8 and 16 dpi. The pathophysiology of Acanthamoeba infection in the skin is complex. The data presented in this paper add new insight, but they are not sufficient to explain the role of the studied receptors and cytokines. The clinical picture and mechanisms of host response appear to be influenced by the route of infection, immunological status and microorganisms carried within the parasites. CA remains a multifactorial phenomenon.
Collapse
Affiliation(s)
- A Wojtkowiak-Giera
- Department of Biology and Medical Parasitology, Poznan University of Medical Sciences, 4 Swiecickiego Street, 60-781, Poznan, Poland
| | - D Kosik-Bogacka
- Department of Biology, Medical and Pharmaceutical Botany, Pomeranian Medical University in Szczecin, 72 Powstancow Wielkopolskich Avenue, 70-111, Szczecin, Poland
| | - N Łanocha-Arendarczyk
- Department of Biology, Medical and Pharmaceutical Botany, Pomeranian Medical University in Szczecin, 72 Powstancow Wielkopolskich Avenue, 70-111, Szczecin, Poland.
| | - A Kolasa
- Department of Histology and Embryology, Pomeranian Medical University in Szczecin, 72 Powstancow Wielkopolskich Avenue, 70-111, Szczecin, Poland
| | - K Kot
- Department of Biology, Medical and Pharmaceutical Botany, Pomeranian Medical University in Szczecin, 72 Powstancow Wielkopolskich Avenue, 70-111, Szczecin, Poland
| | - P Solarczyk
- Department of Biology and Medical Parasitology, Poznan University of Medical Sciences, 4 Swiecickiego Street, 60-781, Poznan, Poland
| | - M Derda
- Department of Biology and Medical Parasitology, Poznan University of Medical Sciences, 4 Swiecickiego Street, 60-781, Poznan, Poland
| |
Collapse
|
2
|
Romero AH, Delgado F. 4-Aminoquinoline as a privileged scaffold for the design of leishmanicidal agents: structure-property relationships and key biological targets. Front Chem 2025; 12:1527946. [PMID: 39981131 PMCID: PMC11841433 DOI: 10.3389/fchem.2024.1527946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 12/26/2024] [Indexed: 02/22/2025] Open
Abstract
Leishmaniasis is one of the most important neglected tropical diseases, with more than two million new cases annually. It is endemic in several regions worldwide, representing a public health problem for more than 88 countries, in particular in the tropical and subtropical regions of developing countries. At the moment, there are neither approved vaccines nor effective drugs for the treatment of human leishmaniasis for any of its three typical clinical manifestations, and, importantly, the drugs of clinical use have several side effects, require complex administration regimens, present high cost, and are ineffective in many populations due to pathogen resistance. Moreover, beyond the pharmacological exigencies, there are other challenges concerning its parasitic nature, such as its great genetic plasticity and adaptability, enabling it to activate a battery of genes to develop resistance quickly. All these aspects demand the identification and development of new, safe, and effective chemical systems, which must not only be focused on medicinal chemistry and pharmacological aspects but also consider key aspects relative to parasite survival. In this sense, the quinolines and, in particular, 4-aminoquinoline, represent a privileged scaffold for the design of potential leishmanicidal candidates due not only to their versatility to generate highly active and selective compounds but also to their correlation with well-defined biological targets. These facts make it possible to generate safe leishmanicidal agents targeted at key aspects of parasite survival. The current review summarizes the most current examples of leishmanicidal agents based on 4-aminoquinolines focusing the analysis on two essential aspects: (i) structure-property relationship to identify the key pharmacophores and (ii) mode of action focused on key targets in parasite survival (e.g., depolarization of potential mitochondrial, accumulation into macrophage lysosome, and immunostimulation of host cells). With that information, we seek to give useful guidelines for interested researchers to face the drug discovery and development process for selective and potent leishmanicidal agents based on 4-aminoquinolines.
Collapse
Affiliation(s)
- Angel H. Romero
- Grupo de Química Orgánica Medicinal, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | | |
Collapse
|
3
|
Roy K, Ghosh S, Karmakar S, Mandal P, Hussain A, Dutta A, Pal C. Inverse correlation between Leishmania-induced TLR1/2 and TGF-β differentially regulates parasite persistence in bone marrow during the chronic phase of infection. Cytokine 2025; 185:156811. [PMID: 39612658 DOI: 10.1016/j.cyto.2024.156811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 12/01/2024]
Abstract
Host-tissue preference is a critical aspect of parasitic infections and is directly correlated with species diversity. Even the same species, Leishmania donovani, infects the host's bone marrow, spleen, and liver differentially. The tissue-specific persistence of Leishmania results from host-pathogen immune conflicts and arguments. The protective pro-host or destructive pro-parasitic role of TLRs during L. donovani infection has been well established, but what entirely missing is the influence of TLRs on tissue-specific parasite persistence. We observed that the parasites induced differential expression of TLR1/2 in the bone marrow but not in the spleen. Interestingly, the rate of Leishmania infection was found to be positively correlated with TLR1/2-mediated upregulation of myelopoietic cytokines, M-CSF, GM-CSF, IL-6, and IL-3, leading to the expansion of Ly6ChiCCR2+ monocytes, however, negatively correlated with the expression of the disease hallmark cytokines, TNF-α, TGF-β, and IL-10, along the course of infection in the bone marrow. Leishmania induced the activation of bone marrow-specific TLR1/2 to promote Ly6ChiCCR2+ monocytes for its safe shelter vis-à-vis infection establishment. Consequently, the established infection initiated the release of TNF-α, TGF-β, and IL-10 in the bone marrow. Post-infection time-kinetic study affirmed that TGF-β had a significant negative influence on the expression of TLR1/2 heterodimer in the bone marrow niche. To the best of our knowledge, this is the first report to show that the inverse correlation of TLR1/2 - TGF-β can be instrumental in tissue-specific parasite persistence during Leishmania infection.
Collapse
Affiliation(s)
- Kamalika Roy
- Cellular Immunology and Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, North 24 Parganas, Barasat, West Bengal, India
| | - Sanhita Ghosh
- Cellular Immunology and Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, North 24 Parganas, Barasat, West Bengal, India
| | - Suman Karmakar
- Cellular Immunology and Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, North 24 Parganas, Barasat, West Bengal, India
| | - Pritam Mandal
- Cellular Immunology and Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, North 24 Parganas, Barasat, West Bengal, India
| | - Aabid Hussain
- Cellular Immunology and Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, North 24 Parganas, Barasat, West Bengal, India
| | - Aritri Dutta
- Cellular Immunology and Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, North 24 Parganas, Barasat, West Bengal, India
| | - Chiranjib Pal
- Cellular Immunology and Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, North 24 Parganas, Barasat, West Bengal, India.
| |
Collapse
|
4
|
Fadaie M, Shahmoradi Z, Khanahmad H. Immunoinformatic approach to the design of a novel multi-epitope vaccine against Leishmania major fused to human IgG-Fc. Res Pharm Sci 2024; 19:729-745. [PMID: 39911897 PMCID: PMC11792711 DOI: 10.4103/rps.rps_145_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/14/2024] [Accepted: 11/18/2024] [Indexed: 02/07/2025] Open
Abstract
Background and purpose Cutaneous leishmaniasis poses significant health and socioeconomic challenges, making vaccine development a top priority, especially in endemic regions. Cysteine proteases, KMP-11, and HASPB proteins are promising candidates for leishmaniasis vaccine development owing to their immunogenic properties and capacity to provoke robust immune responses, as evidenced by different investigations. This study aimed to design a recombinant chimeric protein (MEV-Fc) vaccine using multi-epitopes from these Leishmania major proteins. Experimental approach The antigens were subjected to immunoinformatic prediction and screening of HTL, CTL, and B-cell epitopes. The multi-epitope protein was designed with significantly high-scoring epitopes and suitable linkers. Natural adjuvants were then added to enhance immunogenicity. Vaccine potency was innovatively improved by covalently fusing human IgG1 Fc with multi-epitope protein. To investigate how the MEV-Fc vaccine interacts with Toll-like receptors, molecular docking, multi-scale normal mode analysis simulation, and computational immune simulation were employed to study humoral and cellular immune responses. Findings/Results The results demonstrated the vaccine's antigenicity, stability, and nontoxicity. The structural validation confirmed the accuracy of the 3D models, indicating robust interactions with TLR2 and TLR4, with binding free energies of -1269.9 and -1128.7 (kcal/mol), respectively. Immune simulation results showed significant increases in IgM and IgG antibody levels following three vaccinations, along with enhanced activation of B cells, helper T cells, and cytotoxic T lymphocytes. Conclusion and implications These findings provide novel insights for developing effective candidates for cutaneous leishmaniasis vaccines. However, laboratory experiments are necessary to evaluate its protective effects.
Collapse
Affiliation(s)
- Mahmood Fadaie
- Skin Diseases and Leishmaniasis Research Center, Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zabihollah Shahmoradi
- Skin Diseases and Leishmaniasis Research Center, Department of Dermatology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Khanahmad
- Skin Diseases and Leishmaniasis Research Center, Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
5
|
Ihedioha OC, Marcarian HQ, Sivakoses A, Beverley SM, McMahon-Pratt D, Bothwell ALM. Leishmania major surface components and DKK1 signalling via LRP6 promote migration and longevity of neutrophils in the infection site. Front Immunol 2024; 15:1473133. [PMID: 39502693 PMCID: PMC11534728 DOI: 10.3389/fimmu.2024.1473133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/30/2024] [Indexed: 11/08/2024] Open
Abstract
Background Host-related factors highly regulate the increased circulation of neutrophils during Leishmania infection. Platelet-derived Dickkopf-1 (DKK1) is established as a high-affinity ligand to LRP6. Recently, we demonstrated that DKK1 upregulates leukocyte-platelet aggregation, infiltration of neutrophils to the draining lymph node and Th2 differentiation during Leishmania infection, suggesting the potential involvement of the DKK1-LRP6 signalling pathway in neutrophil migration in infectious diseases. Results In this study, we further explored the potential role of DKK1-LRP6 signalling in the migration and longevity of activated neutrophils in the infection site using BALB/c mice with PMNs deficient in LRP6 (LRP6NKO) or BALB/c mice deficient in both PMN LRP6 and platelet DKK1 (LRP6NKO DKK1PKO). Relative to the infected wild-type BALB/c mice, reduced neutrophil activation at the infection site of LRP6NKO or LRP6NKO DKK1PKO mice was noted. The neutrophils obtained from either infected LRP6NKO or LRP6NKO DKK1PKO mice additionally showed a high level of apoptosis. Notably, the level of LRP6 expressing neutrophils was elevated in infected BALB/c mice. Relative to infected BALB/c mice, a significant reduction in parasite load was observed in both LRP6NKO and LRP6NKO DKK1PKO infected mice. Notably, DKK1 levels were comparable in the LRP6NKO and BALB/c mice in response to infection, indicating that PMN activation is the major pathway for DKK1 in promoting parasitemia. Parasite-specific components also play a crucial role in modulating neutrophil circulation in Leishmania disease. Thus, we further determine the contribution of Leishmania membrane components in the migration of neutrophils to the infection site using null mutants deficient in LPG synthesis (Δlpg1- ) or lacking all ether phospholipids (plasmalogens, LPG, and GIPLs) synthesis (Δads1- ). Relative to the WT controls, Δads1- parasite-infected mice showed a sustained decrease in neutrophils and neutrophil-platelet aggregates (for at least 14 days PI), while neutrophils returned to normal in Δlpg1- parasite-infected mice after day 3 PI. Conclusion Our results suggest that DKK1 signalling and Leishmania pathogen-associated molecular patterns appear to regulate the migration and sustenance of viable activated neutrophils in the infection site resulting in chronic type 2 cell-mediated inflammation.
Collapse
Affiliation(s)
- Olivia C. Ihedioha
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Haley Q. Marcarian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Anutr Sivakoses
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Stephen M. Beverley
- Department of Molecular Microbiology, Washington University School of Medicine in St Louis, St. Louis, MO, United States
| | - Diane McMahon-Pratt
- Department of Epidemiology of Infectious Diseases, Yale School of Public Health, New Haven, CT, United States
| | - Alfred L. M. Bothwell
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
6
|
Lafleur A, Daffis S, Mowbray C, Arana B. Immunotherapeutic Strategies as Potential Treatment Options for Cutaneous Leishmaniasis. Vaccines (Basel) 2024; 12:1179. [PMID: 39460345 PMCID: PMC11511131 DOI: 10.3390/vaccines12101179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Cutaneous leishmaniasis (CL), caused by protozoan parasites of the Leishmania genus, is prevalent in tropical and subtropical regions, with important morbidity, particularly in low- to middle-income countries. Current systemic treatments, including pentavalent antimonials and miltefosine, are associated with significant toxicity, reduced efficacy, and are frequently ineffective in cases of severe or chronic CL. Immunotherapies leverage the immune system to combat microbial infection and offer a promising adjunct or alternative approach to the current standard of care for CL. However, the heterogeneous clinical presentation of CL, which is dependent on parasite species and host immunity, may require informed clinical intervention with immunotherapies. This review explores the clinical and immunological characteristics of CL, emphasising the current landscape of immunotherapies in in vivo models and clinical studies. Such immune-based interventions aim to modulate immune responses against Leishmania, with additive therapeutic effects enabling the efficacy of lower drug doses and decreasing the associated toxicity. Understanding the mechanisms that underlie immunotherapy for CL provides critical insights into developing safer and more effective treatments for this neglected tropical disease. Identifying suitable therapeutic candidates and establishing their safety and efficacy are essential steps in this process. However, the feasibility and utility of these treatments in resource-limited settings must also be considered, taking into account factors such as cost of production, temperature stability, and overall patient access.
Collapse
Affiliation(s)
- Andrea Lafleur
- Doctoral Training Centre, University of Oxford, Oxford OX1 3NP, UK
| | - Stephane Daffis
- Drugs for Neglected Diseases initiative (DNDi), 1202 Geneva, Switzerland; (S.D.)
| | - Charles Mowbray
- Drugs for Neglected Diseases initiative (DNDi), 1202 Geneva, Switzerland; (S.D.)
| | - Byron Arana
- Drugs for Neglected Diseases initiative (DNDi), 1202 Geneva, Switzerland; (S.D.)
| |
Collapse
|
7
|
Goris M, Passelli K, Peyvandi S, Díaz-Varela M, Billion O, Prat-Luri B, Demarco B, Desponds C, Termote M, Iniguez E, Dey S, Malissen B, Kamhawi S, Hurrell BP, Broz P, Tacchini-Cottier F. NLRP1-dependent activation of Gasdermin D in neutrophils controls cutaneous leishmaniasis. PLoS Pathog 2024; 20:e1012527. [PMID: 39250503 PMCID: PMC11412672 DOI: 10.1371/journal.ppat.1012527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 09/19/2024] [Accepted: 08/22/2024] [Indexed: 09/11/2024] Open
Abstract
Intracellular pathogens that replicate in host myeloid cells have devised ways to inhibit the cell's killing machinery. Pyroptosis is one of the host strategies used to reduce the pathogen replicating niche and thereby control its expansion. The intracellular Leishmania parasites can survive and use neutrophils as a silent entry niche, favoring subsequent parasite dissemination into the host. Here, we show that Leishmania mexicana induces NLRP1- and caspase-1-dependent Gasdermin D (GSDMD)-mediated pyroptosis in neutrophils, a process critical to control the parasite-induced pathology. In the absence of GSDMD, we observe an increased number of infected dermal neutrophils two days post-infection. Using adoptive neutrophil transfer in neutropenic mice, we show that pyroptosis contributes to the regulation of the neutrophil niche early after infection. The critical role of neutrophil pyroptosis and its positive influence on the regulation of the disease outcome was further demonstrated following infection of mice with neutrophil-specific deletion of GSDMD. Thus, our study establishes neutrophil pyroptosis as a critical regulator of leishmaniasis pathology.
Collapse
Affiliation(s)
- Michiel Goris
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Katiuska Passelli
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Sanam Peyvandi
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Miriam Díaz-Varela
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Oaklyne Billion
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Borja Prat-Luri
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Benjamin Demarco
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Chantal Desponds
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Manon Termote
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Eva Iniguez
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Somaditya Dey
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
- Post Graduate Department of Zoology, Barasat Government College, Barasat, West Bengal, India
| | - Bernard Malissen
- INSERM, CNRS, Centre D’Immunologie de Marseille-Luminy, Aix-Marseille Université, Marseille, France
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Benjamin P. Hurrell
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Petr Broz
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Fabienne Tacchini-Cottier
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
8
|
Ibañez-Escribano A, Gomez-Muñoz MT, Mateo M, Fonseca-Berzal C, Gomez-Lucia E, Perez RG, Alunda JM, Carrion J. Microbial Matryoshka: Addressing the Relationship between Pathogenic Flagellated Protozoans and Their RNA Viral Endosymbionts (Family Totiviridae). Vet Sci 2024; 11:321. [PMID: 39058005 PMCID: PMC11281412 DOI: 10.3390/vetsci11070321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/12/2024] [Accepted: 07/14/2024] [Indexed: 07/28/2024] Open
Abstract
Three genera of viruses of the family Totiviridae establish endosymbiotic associations with flagellated protozoa responsible for parasitic diseases of great impact in the context of One Health. Giardiavirus, Trichomonasvirus, and Leishmaniavirus infect the protozoa Giardia sp., Trichomonas vaginalis, and Leishmania sp., respectively. In the present work, we review the characteristics of the endosymbiotic relationships established, the advantages, and the consequences caused in mammalian hosts. Among the common characteristics of these double-stranded RNA viruses are that they do not integrate into the host genome, do not follow a lytic cycle, and do not cause cytopathic effects. However, in cases of endosymbiosis between Leishmaniavirus and Leishmania species from the Americas, and between Trichomonasvirus and Trichomonas vaginalis, it seems that it can alter their virulence (degree of pathogenicity). In a mammalian host, due to TLR3 activation of immune cells upon the recognition of viral RNA, uncontrolled inflammatory signaling responses are triggered, increasing pathological damage and the risk of failure of conventional standard treatment. Endosymbiosis with Giardiavirus can cause the loss of intestinal adherence of the protozoan, resulting in a benign disease. The current knowledge about viruses infecting flagellated protozoans is still fragmentary, and more research is required to unravel the intricacies of this three-way relationship. We need to develop early and effective diagnostic methods for further development in the field of translational medicine. Taking advantage of promising biotechnological advances, the aim is to develop ad hoc therapeutic strategies that focus not only on the disease-causing protozoan but also on the virus.
Collapse
Affiliation(s)
- Alexandra Ibañez-Escribano
- Department of Microbiology and Parasitology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (A.I.-E.); (M.M.); (C.F.-B.)
| | - Maria Teresa Gomez-Muñoz
- ICPVet Research Group, Department of Animal Health, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (M.T.G.-M.); (R.G.P.); (J.M.A.)
- Research Institute Hospital 12 de Octubre, 28041 Madrid, Spain
| | - Marta Mateo
- Department of Microbiology and Parasitology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (A.I.-E.); (M.M.); (C.F.-B.)
- ICPVet Research Group, Department of Animal Health, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (M.T.G.-M.); (R.G.P.); (J.M.A.)
- Research Institute Hospital 12 de Octubre, 28041 Madrid, Spain
| | - Cristina Fonseca-Berzal
- Department of Microbiology and Parasitology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (A.I.-E.); (M.M.); (C.F.-B.)
| | - Esperanza Gomez-Lucia
- Animal Viruses Research Group, Department of Animal Health, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain;
| | - Raquel Garcia Perez
- ICPVet Research Group, Department of Animal Health, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (M.T.G.-M.); (R.G.P.); (J.M.A.)
| | - Jose M. Alunda
- ICPVet Research Group, Department of Animal Health, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (M.T.G.-M.); (R.G.P.); (J.M.A.)
- Research Institute Hospital 12 de Octubre, 28041 Madrid, Spain
| | - Javier Carrion
- ICPVet Research Group, Department of Animal Health, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (M.T.G.-M.); (R.G.P.); (J.M.A.)
- Research Institute Hospital 12 de Octubre, 28041 Madrid, Spain
| |
Collapse
|
9
|
Banesh S, Gupta N, Reddy CV, Mallikarjunachari U, Patil N, Uddhavesh S, Saudagar P. A novel approach to design chimeric multi epitope vaccine against Leishmania exploiting infected host cell proteome. Heliyon 2024; 10:e31306. [PMID: 38813178 PMCID: PMC11133825 DOI: 10.1016/j.heliyon.2024.e31306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 05/14/2024] [Accepted: 05/14/2024] [Indexed: 05/31/2024] Open
Abstract
Leishmaniasis is a major infectious disease having high mortality which could be attributed to lack of a suitable vaccine candidate. We propose a novel approach to design multiepitope vaccine to leishmaniasis exploiting specific membrane proteome from infected macrophage from host. The MHC-I, MHC-II and BC epitopes predicted for unique proteins from the infected macrophages and Leishmania and a MEV designed in various combinations (1a-1m). The epitope arrangements 1a, 1k, 1l, and 1 m showed a strong antigenicity profile and immune response. The molecular dynamics simulation indicate the 1k, 1l, and 1 m constructs have strong affinity toward TLR-2, TLR-3, and TLR-4. Overall the structural and immunogenicity profile suggests 1k is top candidate. Further, a computational model system with TLR-2, TLR-3, TLR-4, BCR, MHC-I and MHC-II was generated for 1k construct to understand the MEV interactions with immune components. Dihedral distribution and distance was enumerated to understand the movement of immune components towards 1k. The results indicate 1k has strong affinity for the immune response molecules especially TLR-3, BCR and MHC-II are coming in close contact with the MEV through the simulation. The study suggests that designed multi-epitope vaccine 1k has potential to induce proper immune response but warrants further studies.
Collapse
Affiliation(s)
- Sooram Banesh
- Department of Biotechnology, National Institute of Technology-Warangal, Warangal, 506004, Telangana, India
| | - Neharika Gupta
- Department of Biotechnology, National Institute of Technology-Warangal, Warangal, 506004, Telangana, India
| | - Chethireddy Vihadhar Reddy
- Department of Biotechnology, National Institute of Technology-Warangal, Warangal, 506004, Telangana, India
| | - Uppuladinne Mallikarjunachari
- High Performance Computing - Medical and Bioinformatics Applications, Centre for Development of Advanced Computing (C-DAC), Pune, Maharastra, India
| | - Nupoor Patil
- Department of Biotechnology, National Institute of Technology-Warangal, Warangal, 506004, Telangana, India
| | - Sonavane Uddhavesh
- High Performance Computing - Medical and Bioinformatics Applications, Centre for Development of Advanced Computing (C-DAC), Pune, Maharastra, India
| | - Prakash Saudagar
- Department of Biotechnology, National Institute of Technology-Warangal, Warangal, 506004, Telangana, India
| |
Collapse
|
10
|
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
|
11
|
Zhou X, Li JJ, Li S, Liu HH, Xu DD, Chi CF, Zheng LB. Transcriptomic analysis of large yellow croaker (Larimichthys crocea) reveals the suppression of the inflammatory response from Cryptocaryon irritans infection. FISH & SHELLFISH IMMUNOLOGY 2024; 144:109258. [PMID: 38042226 DOI: 10.1016/j.fsi.2023.109258] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/13/2023] [Accepted: 11/24/2023] [Indexed: 12/04/2023]
Abstract
Large yellow croaker (Larimichthys crocea) is the most productive marine fish in China. Cryptocaryon irritans is an extremely destructive parasite that causes great economic losses in large yellow croaker aquaculture industry. Therefore, it is very necessary to study the immune response of large yellow croaker in response to C. irritans infection. In this study, the transcriptomic profiles of large yellow croaker were sequenced and analyzed in the brain and head kidney at 72 h after C. irritans infection. Cytokines and chemokines related terms were significantly enriched based on the GO enrichment of down-regulated differentially expressed genes (DEGs) from the head kidney. Meanwhile, cytokine-cytokine receptor interaction was significantly enriched based on the KEGG enrichment of up-regulated DEGs from the brain and down-regulated DEGs from the head kidney, respectively. Moreover, the majority of inflammation-related DEGs were significantly up-regulated in the brain, but distinctly down-regulated in the head kidney. These results showed that the brain and head kidney might play different roles against C. irritans infection, and the inflammatory response of large yellow croaker may be restrained during C. irritans infection. Taken together, the transcriptomic analyses will be helpful to more comprehensively understand the immune mechanism of teleost against C. irritans infection, and provide a theoretical basis for the prevention and treatment of Cryptosporidiosis.
Collapse
Affiliation(s)
- Xu Zhou
- National and Provincial Joint Engineering Research Centre for Marine Germplasm Resources Exploration and Utilization, School of Marine Science and Technology, Zhejiang Ocean University, 1st Haidanan Road, Changzhi Island, Lincheng, Zhoushan, 316022, China
| | - Jun-Jie Li
- National and Provincial Joint Engineering Research Centre for Marine Germplasm Resources Exploration and Utilization, School of Marine Science and Technology, Zhejiang Ocean University, 1st Haidanan Road, Changzhi Island, Lincheng, Zhoushan, 316022, China
| | - Shuang Li
- National and Provincial Joint Engineering Research Centre for Marine Germplasm Resources Exploration and Utilization, School of Marine Science and Technology, Zhejiang Ocean University, 1st Haidanan Road, Changzhi Island, Lincheng, Zhoushan, 316022, China
| | - Hui-Hui Liu
- National and Provincial Joint Engineering Research Centre for Marine Germplasm Resources Exploration and Utilization, School of Marine Science and Technology, Zhejiang Ocean University, 1st Haidanan Road, Changzhi Island, Lincheng, Zhoushan, 316022, China
| | - Dong-Dong Xu
- Marine Fishery Institute of Zhejiang Province, Key Lab of Mariculture and Enhancement of Zhejiang Province, Zhoushan, 316100, China
| | - Chang-Feng Chi
- National and Provincial Joint Engineering Research Centre for Marine Germplasm Resources Exploration and Utilization, School of Marine Science and Technology, Zhejiang Ocean University, 1st Haidanan Road, Changzhi Island, Lincheng, Zhoushan, 316022, China.
| | - Li-Bing Zheng
- National and Provincial Joint Engineering Research Centre for Marine Germplasm Resources Exploration and Utilization, School of Marine Science and Technology, Zhejiang Ocean University, 1st Haidanan Road, Changzhi Island, Lincheng, Zhoushan, 316022, China.
| |
Collapse
|
12
|
Ihedioha OC, Sivakoses A, Beverley SM, McMahon-Pratt D, Bothwell ALM. Leishmania major-derived lipophosphoglycan influences the host's early immune response by inducing platelet activation and DKK1 production via TLR1/2. Front Immunol 2023; 14:1257046. [PMID: 37885890 PMCID: PMC10598878 DOI: 10.3389/fimmu.2023.1257046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/25/2023] [Indexed: 10/28/2023] Open
Abstract
Background Platelets are rapidly deployed to infection sites and respond to pathogenic molecules via pattern recognition receptors (TLR, NLRP). Dickkopf1 (DKK1) is a quintessential Wnt antagonist produced by a variety of cell types including platelets, endothelial cells, and is known to modulate pro-inflammatory responses in infectious diseases and cancer. Moreover, DKK1 is critical for forming leukocyte-platelet aggregates and induction of type 2 cell-mediated immune responses. Our previous publication showed activated platelets release DKK1 following Leishmania major recognition. Results Here we probed the role of the key surface virulence glycoconjugate lipophosphoglycan (LPG), on DKK1 production using null mutants deficient in LPG synthesis (Δlpg1- and Δlpg2-). Leishmania-induced DKK1 production was reduced to control levels in the absence of LPG in both mutants and was restored upon re-expression of the cognate LPG1 or LPG2 genes. Furthermore, the formation of leukocyte-platelet aggregates was dependent on LPG. LPG mediated platelet activation and DKK1 production occurs through TLR1/2. Conclusion Thus, LPG is a key virulence factor that induces DKK1 production from activated platelets, and the circulating DKK1 promotes Th2 cell polarization. This suggests that LPG-activated platelets can drive innate and adaptive immune responses to Leishmania infection.
Collapse
Affiliation(s)
- Olivia C. Ihedioha
- 1Department of Immunobiology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Anutr Sivakoses
- 1Department of Immunobiology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Stephen M. Beverley
- Department of Molecular Microbiology, Washington University School of Medicine in St Louis, St. Louis, MI, United States
| | - Diane McMahon-Pratt
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, United States
| | - Alfred L. M. Bothwell
- 1Department of Immunobiology, College of Medicine, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
13
|
Liao X, He J, Wang R, Zhang J, Wei S, Xiao Y, Zhou Q, Zheng X, Zhu Z, Zheng Z, Li J, Zeng Z, Chen D, Chen J. TLR-2 agonist Pam3CSK4 has no therapeutic effect on visceral leishmaniasis in BALB/c mice and may enhance the pathogenesis of the disease. Immunobiology 2023; 228:152725. [PMID: 37562277 DOI: 10.1016/j.imbio.2023.152725] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/13/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023]
Abstract
Most of the existing Leishmania-related research about TLR-2 agonists was focusing on their role as adjuvants in the vaccine, few studied its therapeutic effect. This paper aims to explore the therapeutic effect of TLR-2 agonist Pam3CSK4 on Leishmania-infected mice and the underlying immune molecular mechanisms. In L. donovani-infected BALB/c mice, one group was treated with Pam3CSK4 after infection and the other group was not treated. Normal uninfected mice treated with Pam3CSK4 or untreated were used as controls. Parasite load, hepatic pathology and serum antibodies were detected to assess the severity of the infection. The expression of immune-related genes, spleen lymphocyte subsets and liver RNA-seq were employed to reveal possible molecular mechanisms. The results showed that the liver and spleen parasite load of infected mice in Pam3CSK4 treated and untreated groups had no statistical difference, indicating Pam3CSK4 might have no therapeutic effect on visceral leishmaniasis. Infected mice treated with Pam3CSK4 possessed more hepatic inflammation focus, lower IgG and IgG2a antibody titers, and a lower proportion of spleen CD3+CD4+ T cells. Transcriptome analysis revealed that Th1/Th2 differentiation, NK cells, Th17 cell, complement system and calcium signaling pathways were down-regulated post-treatment of Pam3CSK4. In this study, TLR-2 agonist Pam3CSK4 showed no therapeutic effect on visceral leishmaniasis in BALB/c mice and might enhance the pathogenesis of the disease possibly due to the down-regulation of several immune-related pathways, which can improve our understanding of the role of TLR-2 in both treatment and vaccine development.
Collapse
Affiliation(s)
- Xuechun Liao
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Jinlei He
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Ruanyan Wang
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Jianhui Zhang
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Shulan Wei
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Yuying Xiao
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Qi Zhou
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Xiaoting Zheng
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Zheying Zhu
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Zhiwan Zheng
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China; Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, China
| | - Jiao Li
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China; Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, China
| | - Zheng Zeng
- Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, China; Chong Qing Animal Disease Prevention and Control Center, Chongqing, China
| | - Dali Chen
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China; Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, China.
| | - Jianping Chen
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China; Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, China.
| |
Collapse
|
14
|
Weber JI, Rodrigues AV, Valério-Bolas A, Nunes T, Carvalheiro M, Antunes W, Alexandre-Pires G, da Fonseca IP, Santos-Gomes G. Insights on Host-Parasite Immunomodulation Mediated by Extracellular Vesicles of Cutaneous Leishmania shawi and Leishmania guyanensis. Cells 2023; 12:1101. [PMID: 37190011 PMCID: PMC10137031 DOI: 10.3390/cells12081101] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/23/2023] [Accepted: 04/01/2023] [Indexed: 05/17/2023] Open
Abstract
Leishmaniasis is a parasitic disease caused by different species of Leishmania and transmitted through the bite of sand flies vector. Macrophages (MΦ), the target cells of Leishmania parasites, are phagocytes that play a crucial role in the innate immune microbial defense and are antigen-presenting cells driving the activation of the acquired immune response. Exploring parasite-host communication may be key in restraining parasite dissemination in the host. Extracellular vesicles (EVs) constitute a group of heterogenous cell-derived membranous structures, naturally produced by all cells and with immunomodulatory potential over target cells. This study examined the immunogenic potential of EVs shed by L. shawi and L. guyanensis in MΦ activation by analyzing the dynamics of major histocompatibility complex (MHC), innate immune receptors, and cytokine generation. L. shawi and L. guyanensis EVs were incorporated by MΦ and modulated innate immune receptors, indicating that EVs cargo can be recognized by MΦ sensors. Moreover, EVs induced MΦ to generate a mix of pro- and anti-inflammatory cytokines and favored the expression of MHCI molecules, suggesting that EVs antigens can be present to T cells, activating the acquired immune response of the host. Since nano-sized vesicles can be used as vehicles of immune mediators or immunomodulatory drugs, parasitic EVs can be exploited by bioengineering approaches for the development of efficient prophylactic or therapeutic tools for leishmaniasis.
Collapse
Affiliation(s)
- Juliana Inês Weber
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), Rua da Junqueira 100, 1349-008 Lisboa, Portugal
| | - Armanda Viana Rodrigues
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), Rua da Junqueira 100, 1349-008 Lisboa, Portugal
| | - Ana Valério-Bolas
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), Rua da Junqueira 100, 1349-008 Lisboa, Portugal
| | - Telmo Nunes
- Microscopy Center, Faculty of Sciences, University of Lisbon, Campo Grande, 1749-016 Lisboa, Portugal
| | - Manuela Carvalheiro
- Research Institute for Medicines, iMed, Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - Wilson Antunes
- Unidade Militar Laboratorial de Defesa Biológica e Química (UMLDBQ), 1849-012 Lisboa, Portugal
| | - Graça Alexandre-Pires
- CIISA, Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Av. Universidade Técnica, 1300-477 Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 2825-466 Setúbal, Portugal
| | - Isabel Pereira da Fonseca
- CIISA, Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Av. Universidade Técnica, 1300-477 Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 2825-466 Setúbal, Portugal
| | - Gabriela Santos-Gomes
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), Rua da Junqueira 100, 1349-008 Lisboa, Portugal
| |
Collapse
|
15
|
Bhattacharya P, Gannavaram S, Ismail N, Saxena A, Dagur PK, Akue A, KuKuruga M, Nakhasi HL. Toll-like Receptor-9 (TLR-9) Signaling Is Crucial for Inducing Protective Immunity following Immunization with Genetically Modified Live Attenuated Leishmania Parasites. Pathogens 2023; 12:pathogens12040534. [PMID: 37111420 PMCID: PMC10143410 DOI: 10.3390/pathogens12040534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/16/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023] Open
Abstract
No human vaccine is available for visceral leishmaniasis (VL). Live attenuated centrin gene-deleted L. donovani (LdCen−/−) parasite vaccine has been shown to induce robust innate immunity and provide protection in animal models. Toll-like receptors (TLRs) are expressed in innate immune cells and are essential for the early stages of Leishmania infection. Among TLRs, TLR-9 signaling has been reported to induce host protection during Leishmania infection. Importantly, TLR-9 ligands have been used as immune enhancers for non-live vaccination strategies against leishmaniasis. However, the function of TLR-9 in the generation of a protective immune response in live attenuated Leishmania vaccines remains unknown. In this study, we investigated the function of TLR-9 during LdCen−/− infection and found that it increased the expression of TLR-9 on DCs and macrophages from ear-draining lymph nodes and spleen. The increase in TLR-9 expression resulted in changes in downstream signaling in DCs mediated through signaling protein myeloid differentiation primary response 88 (MyD88), resulting in activation and nuclear translocation of nuclear factor-κB (NF-κB). This process resulted in an increase in the DC’s proinflammatory response, activation, and DC-mediated CD4+T cell proliferation. Further, LdCen−/− immunization in TLR-9−/− mice resulted in a significant loss of protective immunity. Thus, LdCen−/− vaccine naturally activates the TLR-9 signaling pathway to elicit protective immunity against virulent L. donovani challenge.
Collapse
Affiliation(s)
- Parna Bhattacharya
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research Food and Drug Administration, Silver Spring, MD 20993, USA
- Correspondence: (P.B.); (H.L.N.); Tel.: +1-240-402-8209 (H.L.N.)
| | - Sreenivas Gannavaram
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Nevien Ismail
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Ankit Saxena
- Immune Monitoring Shared Resource, Rutgers, Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Pradeep K. Dagur
- Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Adovi Akue
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Mark KuKuruga
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Hira L. Nakhasi
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research Food and Drug Administration, Silver Spring, MD 20993, USA
- Correspondence: (P.B.); (H.L.N.); Tel.: +1-240-402-8209 (H.L.N.)
| |
Collapse
|
16
|
Bhattacharjee M, Banerjee M, Mukherjee A. In silico designing of a novel polyvalent multi-subunit peptide vaccine leveraging cross-immunity against human visceral and cutaneous leishmaniasis: an immunoinformatics-based approach. J Mol Model 2023; 29:99. [PMID: 36928431 PMCID: PMC10018593 DOI: 10.1007/s00894-023-05503-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 03/08/2023] [Indexed: 03/18/2023]
Abstract
CONTEXT Leishmaniasis is a group of vector-borne infectious diseases caused by over 20 pathogenic Leishmania species that are endemic in many tropical and subtropical countries. The emergence of drug-resistant strains, the adverse side effects of anti-Leishmania drugs, and the absence of a preventative vaccination strategy threaten the sensitive population. Recently, many groups of researchers have exploited the field of reverse vaccinology to develop vaccines, focusing chiefly on inducing immunity against either visceral or cutaneous leishmaniasis. METHODS This present work involves retrieving twelve experimentally validated leishmanial antigenic protein sequences from the UniProt database, followed by their antigenicity profiling employing ANTIGENpro and Vaxijen 2.0 servers. MHC-binding epitopes for the same were predicted using both NetCTL 1.2 and SYFPEITHI servers, while epitopes for B cell were computed using ABCpred and BepiPred 2.0 servers. The screened epitopes with significantly higher scores were utilized for designing the vaccine construct with appropriate linkers and natural adjuvant. The secondary and tertiary structures of the synthetic peptide were determined by conditional random fields, shallow neural networks, and profile-profile threading alignment with iterative structure assembly simulations, respectively. The 3-D vaccine model was validated through CASP10-tested refinement and the MolProbity web server. Molecular docking and multi-scale normal mode analysis simulation were performed to analyze the best vaccine-TLR complex. Finally, computational immune simulation findings revealed promising cellular and humoral immune responses, suggesting that the engineered chimeric peptide is a potential broad-spectrum vaccine against visceral and cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Mainak Bhattacharjee
- Department of Biotechnology, Heritage Institute of Technology, 994, Madurdaha, Kolkata, 700107, India
| | - Monojit Banerjee
- Department of Zoology, Triveni Devi Bhalotia College, Raniganj, 713347, India
| | - Arun Mukherjee
- Department of Zoology, Triveni Devi Bhalotia College, Raniganj, 713347, India.
| |
Collapse
|
17
|
Single nucleotide polymorphisms in genes involved in immune responses and outcome of tegumentary leishmaniasis. Acta Trop 2022; 235:106660. [PMID: 35988820 DOI: 10.1016/j.actatropica.2022.106660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 11/24/2022]
Abstract
Leishmaniases are neglected tropical diseases with a broad clinical spectrum. Tegumentary leishmaniasis (TL) is a disease caused by different Leishmania species, transmitted by phlebotomine sand flies and distributed worldwide. TL can present a cutaneous (CL) or mucocutaneous (MCL) clinical form depending on factors inherent to the parasite, the host and the vector. Polymorphisms in the immune response genes are host genetic factors that influence the pathogenesis or control of leishmaniasis. Single nucleotide polymorphisms (SNPs) in immune genes have been evaluated in several countries where leishmaniasis is endemic. In this review, we report studies on SNPs in several immune genes that might be associated with susceptibility or resistance to TL. We summarize studies from around the world and in Brazil, highlight the difficulties of these studies and future analyses needed to enhance our knowledge regarding host genetic factors in TL. Understanding the genetic characteristics of the host that facilitate resistance or susceptibility to leishmaniasis can contribute to the development of immunotherapy schedules for this disease. The current treatment methods are toxic, and no human vaccine is available.
Collapse
|
18
|
Díaz E, Febres A, Giammarresi M, Silva A, Vanegas O, Gomes C, Ponte-Sucre A. G Protein-Coupled Receptors as Potential Intercellular Communication Mediators in Trypanosomatidae. Front Cell Infect Microbiol 2022; 12:812848. [PMID: 35651757 PMCID: PMC9149261 DOI: 10.3389/fcimb.2022.812848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Detection and transduction of environmental signals, constitute a prerequisite for successful parasite invasion; i.e., Leishmania transmission, survival, pathogenesis and disease manifestation and dissemination, with diverse molecules functioning as inter-cellular signaling ligands. Receptors [i.e., G protein-coupled receptors (GPCRs)] and their associated transduction mechanisms, well conserved through evolution, specialize in this function. However, canonical GPCR-related signal transduction systems have not been described in Leishmania, although orthologs, with reduced domains and function, have been identified in Trypanosomatidae. These inter-cellular communication means seem to be essential for multicellular and unicellular organism’s survival. GPCRs are flexible in their molecular architecture and may interact with the so-called receptor activity-modifying proteins (RAMPs), which modulate their function, changing GPCRs pharmacology, acting as chaperones and regulating signaling and/or trafficking in a receptor-dependent manner. In the skin, vasoactive- and neuro- peptides released in response to the noxious stimuli represented by the insect bite may trigger parasite physiological responses, for example, chemotaxis. For instance, in Leishmania (V.) braziliensis, sensory [Substance P, SP, chemoattractant] and autonomic [Vasoactive Intestinal Peptide, VIP, and Neuropeptide Y, NPY, chemorepellent] neuropeptides at physiological levels stimulate in vitro effects on parasite taxis. VIP and NPY chemotactic effects are impaired by their corresponding receptor antagonists, suggesting that the stimulated responses might be mediated by putative GPCRs (with essential conserved receptor domains); the effect of SP is blocked by [(D-Pro 2, D-Trp7,9]-Substance P (10-6 M)] suggesting that it might be mediated by neurokinin-1 transmembrane receptors. Additionally, vasoactive molecules like Calcitonin Gene-Related Peptide [CGRP] and Adrenomedullin [AM], exert a chemorepellent effect and increase the expression of a 24 kDa band recognized in western blot analysis by (human-)-RAMP-2 antibodies. In-silico search oriented towards GPCRs-like receptors and signaling cascades detected a RAMP-2-aligned sequence corresponding to Leishmania folylpolyglutamate synthase and a RAMP-3 aligned protein, a hypothetical Leishmania protein with yet unknown function, suggesting that in Leishmania, CGRP and AM activities may be modulated by RAMP- (-2) and (-3) homologs. The possible presence of proteins and molecules potentially involved in GPCRs cascades, i.e., RAMPs, signpost conservation of ancient signaling systems associated with responses, fundamental for cell survival, (i.e., taxis and migration) and may constitute an open field for description of pharmacophores against Leishmania parasites.
Collapse
Affiliation(s)
- Emilia Díaz
- Laboratory of Molecular Physiology, Institute of Experimental Medicine, School of Medicine Luis Razetti, Faculty of Medicine, Universidad Central de Venezuela, Caracas, Venezuela
| | - Anthony Febres
- Section of Infectious Diseases, Baylor College of Medicine, TX, United States
| | - Michelle Giammarresi
- Laboratory of Molecular Physiology, Institute of Experimental Medicine, School of Medicine Luis Razetti, Faculty of Medicine, Universidad Central de Venezuela, Caracas, Venezuela
| | - Adrian Silva
- Laboratory of Molecular Physiology, Institute of Experimental Medicine, School of Medicine Luis Razetti, Faculty of Medicine, Universidad Central de Venezuela, Caracas, Venezuela
| | - Oriana Vanegas
- Pediatric Gastroenterology, University of Iowa, Iowa City, IA, United States
| | - Carlos Gomes
- Royal Berkshire NHS, Foundation Trust, Light House Lab, Bracknell, United Kingdom
| | - Alicia Ponte-Sucre
- Laboratory of Molecular Physiology, Institute of Experimental Medicine, School of Medicine Luis Razetti, Faculty of Medicine, Universidad Central de Venezuela, Caracas, Venezuela
- Medical Mission Institute, Würzburg, Germany
- *Correspondence: Alicia Ponte-Sucre,
| |
Collapse
|
19
|
Gupta AK, Das S, Kamran M, Ejazi SA, Ali N. The Pathogenicity and Virulence of Leishmania - interplay of virulence factors with host defenses. Virulence 2022; 13:903-935. [PMID: 35531875 PMCID: PMC9154802 DOI: 10.1080/21505594.2022.2074130] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Leishmaniasis is a group of disease caused by the intracellular protozoan parasite of the genus Leishmania. Infection by different species of Leishmania results in various host immune responses, which usually lead to parasite clearance and may also contribute to pathogenesis and, hence, increasing the complexity of the disease. Interestingly, the parasite tends to reside within the unfriendly environment of the macrophages and has evolved various survival strategies to evade or modulate host immune defense. This can be attributed to the array of virulence factors of the vicious parasite, which target important host functioning and machineries. This review encompasses a holistic overview of leishmanial virulence factors, their role in assisting parasite-mediated evasion of host defense weaponries, and modulating epigenetic landscapes of host immune regulatory genes. Furthermore, the review also discusses the diagnostic potential of various leishmanial virulence factors and the advent of immunomodulators as futuristic antileishmanial drug therapy.
Collapse
Affiliation(s)
- Anand Kumar Gupta
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Sonali Das
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Mohd Kamran
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Sarfaraz Ahmad Ejazi
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Nahid Ali
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| |
Collapse
|
20
|
Immune Responses in Leishmaniases: An Overview. Trop Med Infect Dis 2022; 7:tropicalmed7040054. [PMID: 35448829 PMCID: PMC9029249 DOI: 10.3390/tropicalmed7040054] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
Leishmaniasis is a parasitic, widespread, and neglected disease that affects more than 90 countries in the world. More than 20 Leishmania species cause different forms of leishmaniasis that range in severity from cutaneous lesions to systemic infection. The diversity of leishmaniasis forms is due to the species of parasite, vector, environmental and social factors, genetic background, nutritional status, as well as immunocompetence of the host. Here, we discuss the role of the immune system, its molecules, and responses in the establishment, development, and outcome of Leishmaniasis, focusing on innate immune cells and Leishmania major interactions.
Collapse
|
21
|
Seth A, Kar S. Host-directed antileishmanial interventions: Harvesting unripe fruits to reach fruition. Int Rev Immunol 2022; 42:217-236. [PMID: 35275772 DOI: 10.1080/08830185.2022.2047670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Leishmaniasis is an exemplary paradigm of immune evasion, fraught with the perils of limited clinical assistance, escalating costs of treatment and made worse with the lack of suitable vaccine. While drugs remain central to large-scale disease control, the growing emergence of parasite resistance necessitates the need for combination therapy involving host-directed immunological agents. Also, since prolonged disease progression is associated with strong immune suppression of the host, augmentation of host immunity via restoration of the immunoregulatory circuit involving antigen-presenting cells and T-cells, activation of macrophage function and/or CD4+ T helper 1 cell differentiation may serve as an ideal approach to resolve severe cases of leishmaniasis. As such, therapies that embody a synergistic approach that involve direct killing of the parasite in addition to elevating host immunity are likely to pave the way for widespread elimination of leishmaniasis in the future. With this review, we aim to recapitulate the various immunotherapeutic agents found to hold promise in antileishmanial treatment both in vitro and in vivo. These include parasite-specific antigens, dendritic cell-targeted therapy, recombinant inhibitors of various components intrinsic to immune cell signaling and agonists or antagonists to immune cells and cytokines. We also summarize their abilities to direct therapeutic skewing of the host cell-immune response and review their potential to combat the disease either alone, or as adjunct modalities.
Collapse
Affiliation(s)
- Anuradha Seth
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Susanta Kar
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
22
|
Mas A, Martínez-Rodrigo A, Carrión J, Orden JA, Alzate JF, Domínguez-Bernal G, Horcajo P. Transcriptomic Profile of Canine DH82 Macrophages Infected by Leishmania infantum Promastigotes with Different Virulence Behavior. Int J Mol Sci 2022; 23:ijms23031466. [PMID: 35163386 PMCID: PMC8835757 DOI: 10.3390/ijms23031466] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/17/2022] [Accepted: 01/25/2022] [Indexed: 02/07/2023] Open
Abstract
Zoonotic visceral leishmaniosis caused by Leishmania infantum is an endemic disease in the Mediterranean Basin affecting mainly humans and dogs, the main reservoir. The leishmaniosis outbreak declared in the Community of Madrid (Spain) led to a significant increase in human disease incidence without enhancing canine leishmaniosis prevalence, suggesting a better adaptation of the outbreak's isolates by other host species. One of the isolates obtained in the focus, IPER/ES/2012/BOS1FL1 (BOS1FL1), has previously demonstrated a different phenotype than the reference strain MCAN/ES/1996/BCN150 (BCN150), characterized by a lower infectivity when interacting with canine macrophages. Nevertheless, not enough changes in the cell defensive response were found to support their different behavior. Thus, we decided to investigate the molecular mechanisms involved in the interaction of both parasites with DH82 canine macrophages by studying their transcriptomic profiles developed after infection using RNA sequencing. The results showed a common regulation induced by both parasites in the phosphoinositide-3-kinase-protein kinase B/Akt and NOD-like receptor signaling pathways. However, other pathways, such as phagocytosis and signal transduction, including tumor necrosis factor, mitogen-activated kinases and nuclear factor-κB, were only regulated after infection with BOS1FL1. These differences could contribute to the reduced infection ability of the outbreak isolates in canine cells. Our results open a new avenue to investigate the true role of adaptation of L. infantum isolates in their interaction with their different hosts.
Collapse
Affiliation(s)
- Alicia Mas
- INMIVET, Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, 28040 Madrid, Spain; (A.M.); (A.M.-R.); (J.C.); (J.A.O.)
| | - Abel Martínez-Rodrigo
- INMIVET, Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, 28040 Madrid, Spain; (A.M.); (A.M.-R.); (J.C.); (J.A.O.)
| | - Javier Carrión
- INMIVET, Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, 28040 Madrid, Spain; (A.M.); (A.M.-R.); (J.C.); (J.A.O.)
| | - José Antonio Orden
- INMIVET, Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, 28040 Madrid, Spain; (A.M.); (A.M.-R.); (J.C.); (J.A.O.)
| | - Juan F. Alzate
- Centro Nacional de Secuenciación Genómica-CNSG, Facultad de Medicina, Departamento de Microbiología y Parasitología, Universidad de Antioquia, Medellín 050010, Colombia;
| | - Gustavo Domínguez-Bernal
- INMIVET, Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, 28040 Madrid, Spain; (A.M.); (A.M.-R.); (J.C.); (J.A.O.)
- Correspondence: ; Tel.: +34-913943814
| | - Pilar Horcajo
- Animal Health and Zoonoses (SALUVET) Group, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, 28040 Madrid, Spain;
| |
Collapse
|
23
|
Sanz CR, Miró G, Sevane N, Reyes-Palomares A, Dunner S. Modulation of Host Immune Response during Leishmania infantum Natural Infection: A Whole-Transcriptome Analysis of the Popliteal Lymph Nodes in Dogs. Front Immunol 2022; 12:794627. [PMID: 35058931 PMCID: PMC8763708 DOI: 10.3389/fimmu.2021.794627] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/10/2021] [Indexed: 12/21/2022] Open
Abstract
Leishmania infantum, the etiological agent of canine leishmaniosis (CanL) in Europe, was responsible of the largest outbreak of human leishmaniosis in Spain. The parasite infects and survives within myeloid lineage cells, causing a potentially fatal disease if left untreated. The only treatment option relies on chemotherapy, although immunotherapy strategies are being considered as novel approaches to prevent progression of the disease. To this aim, a deeper characterization of the molecular mechanisms behind the immunopathogenesis of leishmaniosis is necessary. Thus, we evaluated, for the first time, the host immune response during L. infantum infection through transcriptome sequencing of the popliteal lymph nodes aspirates of dogs with CanL. Differential expression and weighted gene co-expression network analyses were performed, resulting in the identification of 5,461 differentially expressed genes (DEGs) and four key modules in sick dogs, compared to controls. As expected, defense response was the highest enriched biological process in the DEGs, with six genes related to immune response against pathogens (CHI3L1, SLPI, ACOD1, CCL5, MPO, BPI) included among the ten most expressed genes; and two of the key co-expression modules were associated with regulation of immune response, which also positively correlated with clinical stage and blood monocyte concentration. In particular, sick dogs displayed significant changes in the expression of Th1, Th2, Th17 and Tr1 cytokines (e. g. TNF-α, IFN-γ, IL-21, IL-17, IL-15), markers of T cell and NK cell exhaustion (e. g. LAG3, CD244, Blimp-1, JUN), and B cell, monocyte and macrophage disrupted functionality (e. g. CD40LG, MAPK4, IL-1R, NLRP3, BCMA). In addition, we found an overexpression of XBP1 and some other genes involved in endoplasmic reticulum stress and the IRE1 branch of the unfolded protein response, as well as one co-expression module associated with these processes, which could be induced by L. infantum to prevent host cell apoptosis and modulate inflammation-induced lymphangiogenesis at lymph nodes. Moreover, 21 lncRNAs were differentially expressed in sick dogs, and one key co-expression module was associated with chromatin organization, suggesting that epigenetic mechanisms could also contribute to dampening host immune response during natural L. infantum infection in the lymph nodes of dogs suffering from clinical leishmaniosis.
Collapse
Affiliation(s)
- Carolina R Sanz
- Animal Health Department, Veterinary Faculty, Complutense University of Madrid, Madrid, Spain
| | - Guadalupe Miró
- Animal Health Department, Veterinary Faculty, Complutense University of Madrid, Madrid, Spain
| | - Natalia Sevane
- Department of Animal Production, Veterinary Faculty, Complutense University of Madrid, Madrid, Spain
| | - Armando Reyes-Palomares
- Department of Biochemistry and Molecular Biology, Complutense University of Madrid, Madrid, Spain
| | - Susana Dunner
- Department of Animal Production, Veterinary Faculty, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
24
|
Kaushik D, Granato JT, Macedo GC, Dib PRB, Piplani S, Fung J, da Silva AD, Coimbra ES, Petrovsky N, Salunke DB. Toll-like receptor-7/8 agonist kill Leishmania amazonensis by acting as pro-oxidant and pro-inflammatory agent. J Pharm Pharmacol 2021; 73:1180-1190. [PMID: 33940589 PMCID: PMC8359742 DOI: 10.1093/jpp/rgab063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 03/29/2021] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Evaluation of the anti-Leishmanial activity of imidazoquinoline-based TLR7/8 agonists. METHODS TLR7/8-active imidazoquinolines (2 and 3) were synthesized and assessed for activity against Leishmania amazonensis-intracellular amastigotes using mouse peritoneal macrophages. The production of reactive oxygen species (ROS), nitric oxide (NO) and cytokines was determined in infected and non-infected macrophages. KEY FINDINGS The imidazoquinolines, 2 and 3, were primarily agonists of TLR7 with compound 3 also showing modest TLR8 activity. Docking studies showed them to occupy the same binding pocket on TLR7 and 8 as the known agonists, imiquimod and resiquimod. Compounds 2 and 3 inhibited the growth of L. amazonensis-intracellular amastigotes with the most potent compound (3, IC50 = 5.93 µM) having an IC50 value close to miltefosine (IC50 = 4.05 µM), a known anti-Leishmanial drug. Compound 3 induced macrophages to produce ROS, NO and inflammatory cytokines that likely explain the anti-Leishmanial effects. CONCLUSIONS This study shows that activating TLR7 using compounds 2 or 3 induces anti-Leishmanial activity associated with induction of free radicals and inflammatory cytokines able to kill the parasites. While 2 and 3 had a very narrow cytotoxicity window for macrophages, this identifies the possibility to further develop this chemical scaffold to less cytotoxic TLR7/8 agonist for potential use as anti-Leishmanial drug.
Collapse
Affiliation(s)
- Deepender Kaushik
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh, India
| | - Juliana T Granato
- Departamento de Parasitologia, Microbiologia e Imunologia, I.C.B., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Gilson C Macedo
- Departamento de Parasitologia, Microbiologia e Imunologia, I.C.B., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Paula R B Dib
- Departamento de Parasitologia, Microbiologia e Imunologia, I.C.B., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Sakshi Piplani
- Vaxine Pty Ltd., Warradale, South Australia, Australia
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, Australia
| | - Johnson Fung
- Vaxine Pty Ltd., Warradale, South Australia, Australia
| | - Adilson D da Silva
- Departamento de Química, I.C.E., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Elaine S Coimbra
- Departamento de Parasitologia, Microbiologia e Imunologia, I.C.B., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Nikolai Petrovsky
- Vaxine Pty Ltd., Warradale, South Australia, Australia
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, Australia
| | - Deepak B Salunke
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh, India
- National Interdisciplinary Centre of Vaccine, Immunotherapeutics and Antimicrobials (NICOVIA), Panjab University, Chandigarh, India
| |
Collapse
|
25
|
Serrano-Coll H, Cardona-Castro N, Ramos AP, Llanos-Cuentas A. Innate immune response: ally or enemy in cutaneous leishmaniasis? Pathog Dis 2021; 79:6284792. [PMID: 34037758 DOI: 10.1093/femspd/ftab028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 05/24/2021] [Indexed: 12/27/2022] Open
Abstract
Cutaneous leishmaniasis (CL) is an infectious and neglected disease caused by parasites of the genus Leishmania, which produces a wide spectrum of cutaneous manifestations. CL research has shown that the innate immune activity of cells such as neutrophils, natural killers, macrophages, dendritic cells and the complement system are capable of controlling this infection. However, Leishmania can also modulate the immune activity of these cells to promote its own survival and proliferation at the intracellular level. This review discusses the role of the innate immune response in the control and spread of this infection.
Collapse
Affiliation(s)
- Héctor Serrano-Coll
- Grupo de Investigación en Ciencias de la Educación y de la Salud (ICES), UNISANGIL, San Gil, Colombia.,Instituto Colombiano de Medicina Tropical-Universidad CES, Medellín, Colombia
| | - Nora Cardona-Castro
- Instituto Colombiano de Medicina Tropical-Universidad CES, Medellín, Colombia
| | - Ana Pilar Ramos
- Instituto de Medicina Tropical "Alexander Von von Humboldt", Universidad Peruana Cayetano Heredia, Lima, Perú
| | - Alejandro Llanos-Cuentas
- Instituto de Medicina Tropical "Alexander Von von Humboldt", Universidad Peruana Cayetano Heredia, Lima, Perú
| |
Collapse
|
26
|
Katebi A, Varshochian R, Riazi-Rad F, Ganjalikhani-Hakemi M, Ajdary S. Combinatorial delivery of antigen and TLR agonists via PLGA nanoparticles modulates Leishmania major-infected-macrophages activation. Biomed Pharmacother 2021; 137:111276. [PMID: 33485119 DOI: 10.1016/j.biopha.2021.111276] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/19/2020] [Accepted: 12/31/2020] [Indexed: 02/05/2023] Open
Abstract
Appropriate activation of macrophages is critical for the elimination of Leishmania parasites, which resides in this cell. Some species of Leishmania (L.) fails to stimulate macrophages and establish a chronic infection. To overcome this suppression and induce an innate immune response, the effect of PLGA-encapsulated soluble antigens of Leishmania (SLA) along with agonists of TLR1/2 (Pam3CSK4) and TLR7/8 (R848) nanoparticles (NPs) on activation of L. major-infected-macrophages were investigated and were compared with those of soluble formulations. SLA and R848 were encapsulated into the PLGA, while Pam3CSK4 adsorbed onto the surface of nanoparticles. The kinetics of pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α) and iNOS genes expression were investigated by qPCR over 72 h. The parasite load was also quantified by qPCR. The results indicated that engulfment of L. major promastigotes does not induce any pro-inflammatory cytokines expression by macrophages; however, the infected-cells are capable of responding to the TLRs agonists, and a lesser extent, to the SLA stimulation. Encapsulation resulted in increased strength of the IL-1β, IL-6, TNF-α, and increased and prolonged time of iNOS expression. Also, encapsulation showed the leishmanicidal activity by decreasing parasite load in treated NPs formulations. Among the different combinations of the components, the triple (SLA-R848-Pam3CSK4) forms promoted the highest activation of macrophages, followed by dual SLA-Pam3CSK4 and SLA-R848 NPs. In conclusion, the findings of this study indicate that the addition of SLA in combination with TLR1/2 and TLR7/8 agonists either in NPs or in soluble forms overcome the suppression of L. major-infected macrophages. Moreover, encapsulation increases the strength and duration of the cytokines and iNOS expression, in parallel with decreasing parasite load, suggesting a longer availability or delivery of the NPs into the macrophages. These findings highlight the advantages of particulate therapeutic vaccine formulations.
Collapse
Affiliation(s)
- Asal Katebi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, IR, Iran.
| | - Reyhaneh Varshochian
- Department of Pharmaceutics, Shahid Beheshti University of Medical Sciences, Tehran, IR, Iran; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, IR, Iran.
| | - Farhad Riazi-Rad
- Department of Immunology, Pasteur Institute of Iran, Tehran, IR, Iran.
| | - Mazdak Ganjalikhani-Hakemi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, IR, Iran.
| | - Soheila Ajdary
- Department of Immunology, Pasteur Institute of Iran, Tehran, IR, Iran.
| |
Collapse
|
27
|
Mandal A, Kumar M, Kumar A, Sen A, Das P, Das S. TLR4 and TLR9 polymorphism: Probable role in susceptibility among the population of Bihar for Indian visceral leishmaniasis. Innate Immun 2021; 27:493-500. [PMID: 33910419 PMCID: PMC8504264 DOI: 10.1177/1753425920965658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Genetic variations in the host TLRs genes play an important role in susceptibility and/or resistance to visceral leishmaniasis by altering the host-pathogen interaction. In this study, we investigated the association between polymorphisms of TLR4 (Asp299Gly, Thr399Ile) and TLR-9 (T-1237C), with susceptibility to visceral leishmaniasis. A bi-directional PCR amplification of specific alleles technique was used to characterize the distribution of TLR4 (Asp299Gly and Thr399Ile) and TLR9 (T-1237C) polymorphisms. A total of 60 samples were randomly selected from confirmed visceral leishmaniasis patients and 24 endemic healthy volunteers. The samples were genotyped and allele frequencies were determined. We observed that TLR4 Asp299Gly and Thr399Ile genotypes were more frequent in visceral leishmaniasis patients (10% and 15% respectively) compared to controls (4.2% and 8.3% respectively). However, the differences were not significant in TLR4 Asp299Gly and Thr399Ile alleles and genotypes. In the case of TLR9, we observed the frequency of T1237C genotype was higher in visceral leishmaniasis patients (43.3%) than in healthy controls (33.3%). Statistically significant differences were observed in TLR9 T1237C alleles and genotypes. We concluded that TLR9 T1237C, but not TLR4, gene polymorphisms can be regarded as contributors to visceral leishmaniasis susceptibility among the Indian population of Bihar state.
Collapse
Affiliation(s)
- Abhishek Mandal
- Department of Molecular Biology, Indian Council of Medical Research-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Manish Kumar
- Department of Molecular Biology, Indian Council of Medical Research-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Ashish Kumar
- Department of Biochemistry, Indian Council of Medical Research-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Abhik Sen
- Department of Molecular Biology, Indian Council of Medical Research-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Pradeep Das
- Department of Molecular Biology, Indian Council of Medical Research-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Sushmita Das
- Department of Microbiology, All India Institute of Medical Sciences, Patna, India
| |
Collapse
|
28
|
Kupani M, Pandey RK, Mehrotra S. Neutrophils and Visceral Leishmaniasis: Impact on innate immune response and cross-talks with macrophages and dendritic cells. J Cell Physiol 2020; 236:2255-2267. [PMID: 33345353 DOI: 10.1002/jcp.30029] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 08/16/2020] [Indexed: 12/20/2022]
Abstract
Neutrophils with their array of microbicidal activities are the first innate immune cells to guard against infection. They are also most crucial for the host's initial defense against Leishmania parasites which cause clinically diverse diseases ranging from self-healing cutaneous leishmaniasis (CL) to a more severe visceral form, visceral leishmaniasis (VL). Neutrophils are recruited in large numbers at the infection site after bite of sandfly, which is the vector for the disease. The initial interaction of neutrophils with the parasites may modulate the subsequent innate and adaptive immune responses and hence affect the disease outcome. The purpose of this review is to comprehensively appraise the role of neutrophils during the early stages of Leishmania infection with a focus on the visceral form of the disease. In the past decade, new insights regarding the role of neutrophils in VL have surfaced which have been extensively elaborated in the present review. In addition, since much of the information regarding neutrophil-Leishmania early interaction has accumulated through studies on mouse models of CL, these studies are also revisited. We begin by reviewing the factors which drive the recruitment of neutrophils at the site of injection by the sandfly. We then discuss the studies delineating the molecular mechanisms involved in the uptake of the Leishmania parasite by neutrophils and how the parasite subverts their microbicidal functions. In the end, the interaction of infected neutrophils with macrophages and dendritic cells is summarized.
Collapse
Affiliation(s)
- Manu Kupani
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Rajeev K Pandey
- Research & Development, Thermo Fisher Scientific, Bengaluru, Karnataka, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| |
Collapse
|
29
|
Hussain A, Ghosh S, Roy K, Nath S, Sarkar B, Dutta A, Maji P, Basu S, Paul S, Dey S, Chakraborty K, Raychaudhury B, Acharya K, Ganguly J, Pal C. A mushroom derived 'carbohydrate-fraction' reinstates host-immunity and protects from Leishmania donovani infection. Parasite Immunol 2020; 43:e12806. [PMID: 33131110 DOI: 10.1111/pim.12806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 12/13/2022]
Abstract
The anti-leishmanial effect of the 'carbohydrate-fraction', isolated from an edible mushroom Astraeus hygrometricus, was evaluated against Leishmania donovani infection both in vitro and in vivo. Ahf-Car induced the expression of inducible nitric oxide synthase 2 (iNOS2) and pro-inflammatory cytokines like TNF-α and IL-12, with subsequent downregulation of the anti-inflammatory cytokines as TGF-β and IL-10, in vitro and in vivo along with a remarkable increase in the expressions of IL-6, IL-1β, IFN-γ and IRFs, IRF-7 and IRF-8 in vivo. Ahf-Car also reduced the parasite burden in the spleen and liver dose-dependently with a simultaneous proliferation of Ly6C+ cells in the bone marrow of Leishmania-infected experimental animals. It also increased the monocyte population dose-dependently and the expression of the myeloid transcription factor PU.1, in vivo, which presumably signifies the expansion of protective macrophages. Thus, Ahf-Car might be a potent anti-leishmanial lead with unique and effective adjuvant capacity.
Collapse
Affiliation(s)
- Aabid Hussain
- Cellular Immunology and Experimental Therapeutics Laboratory, Department of Zoology, West Bengal State University, Barasat, India
| | - Sanhita Ghosh
- Cellular Immunology and Experimental Therapeutics Laboratory, Department of Zoology, West Bengal State University, Barasat, India
| | - Kamalika Roy
- Cellular Immunology and Experimental Therapeutics Laboratory, Department of Zoology, West Bengal State University, Barasat, India
| | - Supriya Nath
- Cellular Immunology and Experimental Therapeutics Laboratory, Department of Zoology, West Bengal State University, Barasat, India
| | - Biswajyoti Sarkar
- Cellular Immunology and Experimental Therapeutics Laboratory, Department of Zoology, West Bengal State University, Barasat, India
| | - Aritri Dutta
- Cellular Immunology and Experimental Therapeutics Laboratory, Department of Zoology, West Bengal State University, Barasat, India
| | - Priyankar Maji
- Department of Chemistry, The Indian Institute of Engineering Science and Technology, Shibpur, India
| | - Shibani Basu
- Department of Chemistry, The Indian Institute of Engineering Science and Technology, Shibpur, India
| | - Sharmistha Paul
- Cellular Immunology and Experimental Therapeutics Laboratory, Department of Zoology, West Bengal State University, Barasat, India
| | - Somaditya Dey
- Cellular Immunology and Experimental Therapeutics Laboratory, Department of Zoology, West Bengal State University, Barasat, India
| | | | | | - Krishnendu Acharya
- Molecular and Applied Mycology and Plant Pathology Laboratory, Department of Botany, University of Calcutta, Kolkata, India
| | - Jhuma Ganguly
- Department of Chemistry, The Indian Institute of Engineering Science and Technology, Shibpur, India
| | - Chiranjib Pal
- Cellular Immunology and Experimental Therapeutics Laboratory, Department of Zoology, West Bengal State University, Barasat, India
| |
Collapse
|
30
|
Charoensakulchai S, Bualert L, Manomat J, Mungthin M, Leelayoova S, Tan-Ariya P, Siripattanapipong S, Naaglor T, Piyaraj P. Risk Factors of Leishmania Infection among HIV-Infected Patients in Trang Province, Southern Thailand: A Study on Three Prevalent Species. Am J Trop Med Hyg 2020; 103:1502-1509. [PMID: 32700674 DOI: 10.4269/ajtmh.20-0332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
There are two main species of Leishmania reported in Thailand, that is, Leishmania siamensis and Leishmania martiniquensis. Moreover, leishmaniasis cases caused by Leishmania donovani complex were also reported. There is still a lack of information concerning risk factors of Leishmania infection in Thailand. This study aimed to identify the risk factors of Leishmania infection caused by these three species among HIV-infected patients. A cross-sectional study was conducted in HIV clinic at Trang Hospital, Thailand. Nested PCR and sequencing were performed to detect Leishmania DNA in blood and saliva samples and identify Leishmania species. A standardized questionnaire was used to interview individuals. A total of 526 patients were recruited in this study. Sixty-three (12.0%) were positive for L. siamensis, 24 (4.6%) were positive for L. martiniquensis, and 23 (4.4%) were positive for L. donovani complex. Risk factors of L. siamensis infection included using intravenous drug (adjusted odds ratio [AOR] 2.01, 95% CI: 1.01-4.02). Risk factors of L. martiniquensis infection included female gender (AOR 4.23, 95% CI: 1.52-11.75), using recreational drug (AOR 3.43, 95% CI: 1.00-11.74), and having comorbidities (AOR 4.94, 95% CI: 2.00-12.21). Risk factors of L. donovani complex infection included having opportunistic infection (AOR 4.22, 95% CI: 1.00-17.79), CD4 count 200-500 cells/mm3 (AOR 3.64, 95% CI: 1.14-6.86), and not using insect repellent (AOR 3.04, 95% CI: 1.08-8.58). This study identified the risk factors of Leishmania infection caused by three Leishmania species in Thailand. The data could be useful for disease prevention and control. Further studies on trends of Leishmania infection and preventive measures are recommended.
Collapse
Affiliation(s)
| | | | - Jipada Manomat
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Mathirut Mungthin
- Department of Parasitology, Phramongkutklao College of Medicine, Bangkok, Thailand
| | - Saovanee Leelayoova
- Department of Parasitology, Phramongkutklao College of Medicine, Bangkok, Thailand
| | - Peerapan Tan-Ariya
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | - Tawee Naaglor
- Department of Parasitology, Phramongkutklao College of Medicine, Bangkok, Thailand
| | - Phunlerd Piyaraj
- Department of Parasitology, Phramongkutklao College of Medicine, Bangkok, Thailand
| |
Collapse
|
31
|
Reis NFDC, Dupin TV, Costa CR, Toledo MDS, de Oliveira VC, Popi AF, Torrecilhas AC, Xander P. Leishmania amazonensis Promastigotes or Extracellular Vesicles Modulate B-1 Cell Activation and Differentiation. Front Cell Infect Microbiol 2020; 10:573813. [PMID: 33194814 PMCID: PMC7662559 DOI: 10.3389/fcimb.2020.573813] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
B-1 cells are considered an innate-like B cell population that participates in effective innate and adaptive responses to pathogens. B-1 cells produce immunoglobulins, cytokines, chemokines, migrate to inflammatory sites, and differentiate into mononuclear phagocyte-like cells. Murine B-1 cells phagocytosed Leishmaniain vitro and in vivo and participate in immunity against Leishmania. Our group showed that B-1 cells or their extracellular vesicles (EVs) led to a resistance to experimental infection by L. amazonensis. However, the B-1 cells’ responses to Leishmania or EVs isolated from parasites are still poorly characterized. Studying the activation and differentiation of B-1 cells in vivo can contribute to a better understanding of how these cells participate in immunity to L. amazonensis. Thus, we evaluated the expression of myeloid (M-csfr, G-csfr, Spi-1) and lymphoid (EBF, E2A, IL-7R) lineage commitment factors, Toll-like receptors (TLRs), activation cell surface markers, nitric oxide (NO) and reactive oxygen species (ROS) production in murine peritoneal B-1 cells collected after 24 or 48 h post-infection with Leishmania (Leishmania) amazonensis promastigotes or EVs released by the parasites. Our results demonstrated that L. amazonensis infection did not stimulate the expression of CD40, CD80, CD86, F4/80, and MHC II in B-1 cells, but a significant decrease in the production of NO and ROS was observed. The infection induced a significantly higher arginase expression in B-1 cells, but the stimulation with EVs led to a decrease in this gene expression. TLR-2 and TLR-6 had significantly higher expression in B-1 cells from mice intraperitoneally stimulated with the parasite. The TLR-9 expression was higher in animals infected or stimulated for 48 h with EVs. Interestingly, in B-1 cells the stimulus with L. amazonensis led to a substantial increase in the expression of myeloid restricted transcription factors. Thus, our study suggests that the parasites or EVs differently modulated the activation and differentiation of B-1 cells.
Collapse
Affiliation(s)
- Natasha Ferraz de Campos Reis
- Laboratory of Cellular Immunology and Biochemistry of Fungi and Protozoa, Department of Pharmaceutical Sciences, Federal University of São Paulo, São Paulo, Brazil
| | - Talita Vieira Dupin
- Laboratory of Cellular Immunology and Biochemistry of Fungi and Protozoa, Department of Pharmaceutical Sciences, Federal University of São Paulo, São Paulo, Brazil
| | - Carolina Rizzaro Costa
- Laboratory of Cellular Immunology and Biochemistry of Fungi and Protozoa, Department of Pharmaceutical Sciences, Federal University of São Paulo, São Paulo, Brazil
| | - Maytê Dos Santos Toledo
- Laboratory of Cellular Immunology and Biochemistry of Fungi and Protozoa, Department of Pharmaceutical Sciences, Federal University of São Paulo, São Paulo, Brazil
| | - Vivian Cristina de Oliveira
- Department of Microbiology, Immunology and Parasitology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Ana Flavia Popi
- Department of Microbiology, Immunology and Parasitology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Ana Claudia Torrecilhas
- Laboratory of Cellular Immunology and Biochemistry of Fungi and Protozoa, Department of Pharmaceutical Sciences, Federal University of São Paulo, São Paulo, Brazil
| | - Patricia Xander
- Laboratory of Cellular Immunology and Biochemistry of Fungi and Protozoa, Department of Pharmaceutical Sciences, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
32
|
Yadav S, Prakash J, Shukla H, Das KC, Tripathi T, Dubey VK. Design of a multi-epitope subunit vaccine for immune-protection against Leishmania parasite. Pathog Glob Health 2020; 114:471-481. [PMID: 33161887 DOI: 10.1080/20477724.2020.1842976] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Visceral Leishmaniasis (VL) is an insect-borne neglected disease caused by the protozoan parasite Leishmania donovani. In the absence of a commercial vaccine against VL, chemotherapy is currently the only option used for the treatment of VL. Vaccination has been considered as the most effective and powerful tool for complete eradication and control of infectious diseases. In this study, we aimed to design a peptide-based vaccine against L. donovani using immuno-bioinformatic tools. We identified 6 HTL, 18 CTL, and 25 B-cell epitopes from three hypothetical membrane proteins of L. donovani. All these epitopes were used to make a vaccine construct along with linkers. An adjuvant was also added at the N-terminal to enhance its immunogenicity. After that, we checked the quality of this vaccine construct and found that it is nontoxic, nonallergic, and thermally stable. A 3D structure of the vaccine construct was also generated by homology modeling to evaluate its interaction with innate immune receptors (TLR). Molecular docking was performed, which confirmed its binding with a toll-like receptor-2 (TLR-2). The stability of vaccine-TLR-2 complex and underlying interactions were evaluated using molecular dynamic simulation. Lastly, we carried out in silico cloning to check the expression of the final designed vaccine. The designed vaccine construct needs further experimental and clinical investigations to develop it as a safe and effective vaccine against VL infection.
Collapse
Affiliation(s)
- Sunita Yadav
- School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi , Varanasi, India
| | - Jay Prakash
- School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi , Varanasi, India
| | - Harish Shukla
- Molecular and Structural Biophysics Laboratory, Department of Biochemistry, North-EasternHill University , Shillong, India
| | - Kanhu Charan Das
- Molecular and Structural Biophysics Laboratory, Department of Biochemistry, North-EasternHill University , Shillong, India
| | - Timir Tripathi
- Molecular and Structural Biophysics Laboratory, Department of Biochemistry, North-EasternHill University , Shillong, India
| | - Vikash Kumar Dubey
- School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi , Varanasi, India
| |
Collapse
|
33
|
Mas A, Martínez-Rodrigo A, Orden JA, Viñals LM, Domínguez-Bernal G, Carrión J. A further investigation of the leishmaniosis outbreak in Madrid (Spain): low-infectivity phenotype of the Leishmania infantum BOS1FL1 isolate to establish infection in canine cells. Vet Immunol Immunopathol 2020; 230:110148. [PMID: 33189999 DOI: 10.1016/j.vetimm.2020.110148] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 10/28/2020] [Accepted: 11/03/2020] [Indexed: 10/23/2022]
Abstract
Human leishmaniosis caused by Leishmania infantum is a zoonotic disease, with dogs as the main reservoir in Mediterranean Basin countries. The largest European outbreak of human leishmaniosis declared in the southwestern Madrid region (Spain) is characterized by unusual epidemiological and clinical features, such as the emergence of new wild reservoirs (hares and rabbits), whereas the seroprevalence, infection, and severity of canine leishmaniosis have not substantially changed since the first studies conducted in Madrid before the outbreak. Previous studies reported that L. infantum isolates from the Madrid leishmaniosis focus displayed elevated virulence in in vivo models of infection and increased infectivity in murine target cells. With the aim of studying whether changes in the host-parasite interaction and virulence profile have developed, we first assessed the behaviour of one circulating isolate of the outbreak, IPER/ES/2012/BOS1FL1 (BOS1FL1), compared to that of a well-characterized strain from canine leishmaniosis, MCAN/ES/1996/BCN150 (BCN150), in terms of infection capacity (percentage of infected cells, representing infectivity, and number of amastigotes per infected cell, representing the intensity of infection) in canine monocytes and macrophages. BCN150 displayed significantly higher infectivity (76.82 ± 4.40 vs 38.58 ± 2.19; P < 0.0001) and intensity of infection (3.64 ± 0.13 vs 1.83 ± 0.12; P < 0.0001) than BOS1FL1 when interacting with canine cells. Our ROS induction results did not differ significantly between the two isolates or with the responses previously described for other L. infantum isolates. Paradoxically, increased resilience to hydrogen peroxide exposure was observed for BOS1FL1 (% viability 40.62 ± 5.54 vs 26.37 ± 2.93; P = 0.039). Finally, we demonstrated that a decreased intracellular load of BOS1FL1 was associated with increased IFN-γ (261.21 ± 26.29 vs 69.80 ± 9.02; P = 0.0151) and decreased IL-10 production (165.06 ± 23.87 vs 264.41 ± 30.58; P = 0.0002). In this study, we provide the first detailed insight into the differences between the isolate BOS1FL1 from the outbreak in Madrid and the well-characterized strain BCN150 MON-1 obtained from a dog in their response to interacting with canine cells. However, further studies are necessary to shed light on the immune mechanisms resulting in BOS1FL1 exhibiting less virulent behaviour in canine cells than in cells derived from other host species.
Collapse
Affiliation(s)
- Alicia Mas
- INMIVET, Department of Animal Health, School of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Abel Martínez-Rodrigo
- INMIVET, Department of Animal Health, School of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Jose Antonio Orden
- INMIVET, Department of Animal Health, School of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | | | - Gustavo Domínguez-Bernal
- INMIVET, Department of Animal Health, School of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain.
| | - Javier Carrión
- INMIVET, Department of Animal Health, School of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
34
|
Cytokine saga in visceral leishmaniasis. Cytokine 2020; 147:155322. [PMID: 33127259 DOI: 10.1016/j.cyto.2020.155322] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/21/2020] [Accepted: 09/25/2020] [Indexed: 12/12/2022]
Abstract
In humans, infection with Leishmania manifests into a spectrum of diseases. The manifestation of the diseases depend on the resultant evasion of the parasite to immune responses namely by macrophages, which is an exclusive host of Leishmania. The B cells valiantly mount antibody responses, however, to no avail as the Leishmania parasites occupy the intracellular niches of the macrophages and subvert the immune response. Extensive studies have been documented on the role of cell-mediated immunity (CMI) in protection and counter survival strategies of the parasites leading to downregulation of CMI. The present review attempts to discuss the cytokines in progression or resolution of visceral form of leishmaniasis or kala-azar, predominantly affecting the Indian subcontinent. The components/cytokine(s) responsible for the regulation of the critical balance of T helper cells and their subsets have been discussed in the perspective. Therefore, any strategy involving the treatment of visceral leishmania (VL) needs to consider the balance and regulation of T cell function.
Collapse
|
35
|
Sarmah P, Bharali R, Khatonier R, Khan A. Polymorphism in Toll interacting protein (TOLLIP) gene and its association with Visceral Leishmaniasis. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
36
|
Rocha MI, Dias F, Resende M, Sousa M, Duarte M, Tomás AM, Castro H. Leishmania infantum Enhances Migration of Macrophages via a Phosphoinositide 3-Kinase γ-Dependent Pathway. ACS Infect Dis 2020; 6:1643-1649. [PMID: 32369694 DOI: 10.1021/acsinfecdis.0c00080] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Leishmania infantum (L. infantum) and Leishmania major (L. major) are phylogenetically related protozoan parasites that cause different pathologies in humans (visceral and cutaneous infections, respectively). Here, we report on how these obligatory intracellular pathogens differentially affect the migration of macrophages. Resorting to gap closure assays of infected murine bone marrow derived macrophages, we observed that L. infantum enhances the mobility of these cells. This is not the case of L. major, whose impact on macrophage migration is null. Resorting to kinase inhibition assays, we witnessed that chemical inhibition of phosphoinositide 3-kinase-γ (PI3Kγ) critically impairs cell mobility in all experimental conditions. Importantly, the blockade of tyrosine kinases with dasatinib also slows down naı̈ve and L. major-parasitized cells but not macrophages exposed to L. infantum. The dasatinib-resistant phenotype of L. infantum-infected macrophages aligns with the hypothesis that this parasite invokes a tyrosine kinase-independent pathway to increase the PI3Kγ activity of macrophages and enhance migration.
Collapse
Affiliation(s)
- Maria Inês Rocha
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Filipa Dias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Mariana Resende
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Mafalda Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Margarida Duarte
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Ana Maria Tomás
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Helena Castro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| |
Collapse
|
37
|
Chen D, Zhao Y, Feng Y, Jin C, Yang Q, Qiu H, Xie H, Xie S, Zhou Y, Huang J. Expression of TLR2, TLR3, TLR4, and TLR7 on pulmonary lymphocytes of Schistosoma japonicum-infected C57BL/6 mice. Innate Immun 2020; 25:224-234. [PMID: 31018808 PMCID: PMC6830883 DOI: 10.1177/1753425919840424] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Despite the paramount role of TLRs in the induction of innate immune and
inflammatory responses, there is a paucity of studies on the role of TLRs in
Schistosoma japonicum infection. Here, we observed obvious
infiltration of inflammatory cells in S. japonicum-infected
C57BL/6 mouse lungs. Expression and release of IFN-γ, IL-4, and IL-17 were
significantly higher in pulmonary lymphocytes from infected mice compared with
control mice in response to anti-CD3 plus anti-CD28 mAbs. Higher percentages of
TLR2, TLR3, TLR4, and TLR7 were expressed on such lymphocytes, and the TLR
agonists PGN, Poly I:C, LPS, and R848 induced a higher level of IFN-γ. However,
a higher level of IL-4 was found in the supernatant of pulmonary lymphocytes
from infected mice stimulated by these TLR agonists plus CD3 Ab. Only R848 plus
anti-CD3 mAb could induce a higher level of IFN-γ in such lymphocytes. TLR
expressions were then compared on different pulmonary lymphocytes after
infection, including T cells, B cells, NK cells, NKT cells, and γδT cells. The
expression levels of TLR3 on T cells, B cells, NK cells, and γδT cells were
increased in the lungs after infection. NK cells also expressed higher levels of
TLR4 after infection of control mice. Collectively, these findings highlight the
potential role of TLR expression in the context of S. japonicum
infection.
Collapse
Affiliation(s)
- Dianhui Chen
- 1 The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou Medical University, China
| | - Yi Zhao
- 2 Sino-French Hoffmann Institute, School of Basic Medical Sciences and Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, China
| | - Yuanfa Feng
- 2 Sino-French Hoffmann Institute, School of Basic Medical Sciences and Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, China
| | - Chenxi Jin
- 2 Sino-French Hoffmann Institute, School of Basic Medical Sciences and Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, China
| | - Quan Yang
- 2 Sino-French Hoffmann Institute, School of Basic Medical Sciences and Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, China
| | - Huaina Qiu
- 2 Sino-French Hoffmann Institute, School of Basic Medical Sciences and Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, China
| | - Hongyan Xie
- 2 Sino-French Hoffmann Institute, School of Basic Medical Sciences and Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, China
| | - Sihao Xie
- 2 Sino-French Hoffmann Institute, School of Basic Medical Sciences and Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, China
| | - Yi Zhou
- 3 College of Pharmacy, Guangzhou Medical University, China
| | - Jun Huang
- 2 Sino-French Hoffmann Institute, School of Basic Medical Sciences and Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, China
| |
Collapse
|
38
|
Hurdayal R, Nieuwenhuizen NE, Khutlang R, Brombacher F. Inflammatory Dendritic Cells, Regulated by IL-4 Receptor Alpha Signaling, Control Replication, and Dissemination of Leishmania major in Mice. Front Cell Infect Microbiol 2020; 9:479. [PMID: 32039054 PMCID: PMC6992597 DOI: 10.3389/fcimb.2019.00479] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/27/2019] [Indexed: 11/16/2022] Open
Abstract
Leishmaniasis is a vector-borne disease caused by Leishmania parasites. Macrophages are considered the primary parasite host cell, but dendritic cells (DCs) play a critical role in initiating adaptive immunity and controlling Leishmania infection. Accordingly, our previous study in CD11ccreIL-4Rα−/lox mice, which have impaired IL-4 receptor alpha (IL-4Rα) expression on CD11c+ cells including DCs, confirmed a protective role for IL-4/IL-13-responsive DCs in replication and dissemination of parasites during cutaneous leishmaniasis. However, it was unclear which DC subset/s was executing this function. To investigate this, we infected CD11ccreIL-4Rα−/lox and control mice with L. major GFP+ parasites and identified subsets of infected DCs by flow cytometry. Three days after infection, CD11b+ DCs and CD103+ DCs were the main infected DC subsets in the footpad and draining lymph node, respectively and by 4 weeks post-infection, Ly6C+ and Ly6C− CD11b+ DCs were the main infected DC populations in both the lymph nodes and footpads. Interestingly, Ly6C+CD11b+ inflammatory monocyte-derived DCs but not Ly6C−CD11b+ DCs hosted parasites in the spleen. Importantly, intracellular parasitism was significantly higher in IL-4Rα-deficient DCs. In terms of DC effector function, we found no change in the expression of pattern-recognition receptors (TLR4 and TLR9) nor in expression of the co-stimulatory marker, CD80, but MHCII expression was lower in CD11ccreIL-4Rα−/lox mice at later time-points compared to the controls. Interestingly, in CD11ccreIL-4Rα−/lox mice, which have reduced Th1 responses, CD11b+ DCs had impaired iNOS production, suggesting that DC IL-4Rα expression and NO production is important for controlling parasite numbers and preventing dissemination. Expression of the alternative activation marker arginase was unchanged in CD11b+ DCs in CD11creIL-4Rα−/lox mice compared to littermate controls, but RELM-α was upregulated, suggesting IL-4Rα-independent alternative activation. In summary, L. major parasites may use Ly6C+CD11b+ inflammatory DCs derived from monocytes recruited to infection as “Trojan horses” to migrate to secondary lymphoid organs and peripheral sites, and DC IL-4Rα expression is important for controlling infection.
Collapse
Affiliation(s)
- Ramona Hurdayal
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa.,International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Faculty of Health Sciences, South African Medical Research Council on Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Natalie Eva Nieuwenhuizen
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Faculty of Health Sciences, South African Medical Research Council on Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Rethabile Khutlang
- Identity Authentication Research Group, Defence and Security, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - Frank Brombacher
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Faculty of Health Sciences, South African Medical Research Council on Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
39
|
Saha G, Khamar BM, Prerna K, Kumar M, Dubey VK. BLIMP-1 Plays Important Role in the Regulation of Macrophage Pyroptosis for the Growth and Multiplication of Leishmania donovani. ACS Infect Dis 2019; 5:2087-2095. [PMID: 31618572 DOI: 10.1021/acsinfecdis.9b00186] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Visceral leishmaniasis, one of the fatal forms of the disease, is caused by Leishmania donovani and presents morbid clinical manifestations. The parasite evades pro-inflammatory immune responses by several reported mechanisms and modulates the host immune system to cause fatal symptoms. A plethora of reports related to the role of BLIMP-1 and its involvement in suppressing the immune response in various infectious diseases have been documented. Higher parasitic burden due to increased BLIMP-1 production has been reported earlier for malaria and leishmaniasis with no detailed information. We report for the first time the role of BLIMP-1 in suppressing macrophage pyroptosis during L. donovani infection and thereby tweaking the tight regulation of the NFκβ-NLRP3 signaling pathway. Expression analyses of BLIMP-1 and NFκβ have been measured using real-time PCR and Western blotting. The importance of BLIMP-1 has been validated using a siRNA-mediated experiment along with caspase 1 activity, LDH release assay, and infectivity index analyses. An inverse relationship between BLIMP-1 and NFκβ expression has been highlighted during L. donovani infection, which is reversed in blimp-1 deficient cells infected with promastigotes. The above fact has been further validated with caspase 1 activity assay, and LDH release along with IFNγ and TNF-α release assay. Finally, resumption of pyroptosis has been concluded in infected blimp-1 deficient cells in contrast to wild type infected cells. We conjecture that parasites modulate the NFκβ-NLRP3 signaling pathway by taking advantage of BLIMP-1 dependent IL-10 production and finally disrupting an inflammation-mediated pyroptosis cell death pathway in infected cells.
Collapse
Affiliation(s)
- Gundappa Saha
- Department of Biosciences & Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | | | - Kumari Prerna
- School of Biochemical Engineering, Indian Institute of Technology BHU, Varanasi, Uttar Pradesh 221005, India
| | - Manish Kumar
- Department of Biosciences & Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Vikash Kumar Dubey
- Department of Biosciences & Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
- School of Biochemical Engineering, Indian Institute of Technology BHU, Varanasi, Uttar Pradesh 221005, India
| |
Collapse
|
40
|
Yazdani HO, Roy E, Comerci AJ, van der Windt DJ, Zhang H, Huang H, Loughran P, Shiva S, Geller DA, Bartlett DL, Tsung A, Sheng T, Simmons RL, Tohme S. Neutrophil Extracellular Traps Drive Mitochondrial Homeostasis in Tumors to Augment Growth. Cancer Res 2019; 79:5626-5639. [PMID: 31519688 PMCID: PMC6825588 DOI: 10.1158/0008-5472.can-19-0800] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 07/31/2019] [Accepted: 09/09/2019] [Indexed: 12/29/2022]
Abstract
Neutrophil infiltration and neutrophil extracellular traps (NET) in solid cancers are associated with poorer prognosis, but the mechanisms are incompletely understood. We hypothesized that NETs enhance mitochondrial function in tumor cells, providing extra energy for accelerated growth. Metastatic colorectal cancer tissue showed increased intratumoral NETs and supranormal preoperative serum MPO-DNA, a NET marker. Higher MPO-DNA correlated with shorter survival. In mice, subcutaneous tumor implants and hepatic metastases grew slowly in PAD4-KO mice, genetically incapable of NETosis. In parallel experiments, human cancer cell lines grew slower in nu/nu mice treated with DNAse, which disassembles NETs. PAD4-KO tumors manifested decreased proliferation, increased apoptosis, and increased evidence of oxidative stress. PAD4-KO tumors had decreased mitochondrial density, mitochondrial DNA, a lesser degree of ATP production, along with significantly decreased mitochondrial biogenesis proteins PGC1α, TFAM, and NRF-1. In vitro, cancer cells treated with NETs upregulated mitochondrial biogenesis-associated genes, increased mitochondrial density, increased ATP production, enhanced the percentage of cancer cells with reduced mitochondrial membrane potential, and increased the oxygen consumption rate. Furthermore, NETs increased cancer cells' expression of fission and fusion-associated proteins, DRP-1 and MFN-2, and mitophagy-linked proteins, PINK1 and Parkin. All of which were decreased in PAD4-KO tumors. Mechanistically, neutrophil elastase released from NETs activated TLR4 on cancer cells, leading to PGC1α upregulation, increased mitochondrial biogenesis, and accelerated growth. Taken together, NETs can directly alter the metabolic programming of cancer cells to increase tumor growth. NETs represent a promising therapeutic target to halt cancer progression. SIGNIFICANCE: Neutrophils through the release of NETs facilitate the growth of stressed cancer cells by altering their bioenergetics, the inhibition of which induces cell death.
Collapse
Affiliation(s)
- Hamza O Yazdani
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Eva Roy
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | | | - Hongji Zhang
- Department of Surgery, Ohio State University, Wexner Medical Center, Columbus, Ohio
| | - Hai Huang
- Department of Surgery, Ohio State University, Wexner Medical Center, Columbus, Ohio
| | - Patricia Loughran
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for Biologic Imaging, Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sruti Shiva
- Department of Pharmacology and Chemical Biology, Vascular Medicine Institute, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - David A Geller
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - David L Bartlett
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Allan Tsung
- Department of Surgery, Ohio State University, Wexner Medical Center, Columbus, Ohio
| | - Tai Sheng
- Department of Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Richard L Simmons
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Samer Tohme
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
41
|
Silveira FT. What makes mucosal and anergic diffuse cutaneous leishmaniases so clinically and immunopathogically different? A review in Brazil. Trans R Soc Trop Med Hyg 2019; 113:505-516. [PMID: 31140559 DOI: 10.1093/trstmh/trz037] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 03/21/2019] [Accepted: 04/22/2019] [Indexed: 11/14/2022] Open
Abstract
American cutaneous leishmaniasis (ACL) is a parasitic protozoan disease caused by different Leishmania species widely distributed throughout Latin America. Fifteen Leishmania species belonging to the subgenera Viannia, Leishmania and Mundinia are known to cause ACL. Seven of these species are found in Brazil, of which Leishmania (Viannia) braziliensis and Leishmania (Leishmania) amazonensis have the highest potential to cause mucosal (ML) and anergic diffuse cutaneous leishmaniasis (DCL), respectively, the most severe forms of ACL. The clinical and immunopathological differences between these two clinical forms are reviewed here, taking into account their different physiopathogenic mechanisms of dissemination from cutaneous lesions to mucosal tissues in the case of ML and to almost all body surfaces in the case of anergic DCL. We also discuss some immunopathogenic mechanisms of species-specific Leishmania antigens (from the subgenera Viannia and Leishmania) that are most likely associated with the clinical and immunopathological differences between ML and anergic DCL. Those discussions emphasize the pivotal importance of some surface antigens of L. (V.) braziliensis and L. (L.) amazonensis, such as lipophosphoglycan, phosphatidylserine and CD200 (an immunoregulatory molecule that inhibits macrophage activation), that have been shown to exert strong influences on the clinical and immunopathological differences between ML and anergic DCL.
Collapse
Affiliation(s)
- Fernando T Silveira
- Leishmaniasis Laboratory Prof. Dr. Ralph Lainson, Parasitology Department, Evandro Chagas Institute, Rod. BR 316-KM 07, Levilândia, Ananindeua, Pará State, Brazil
- Nucleus of Tropical Medicine, Federal University of Pará, Belém, Pará State, Brazil
| |
Collapse
|
42
|
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
|
43
|
Dias BT, Dias-Teixeira KL, Godinho JP, Faria MS, Calegari-Silva T, Mukhtar MM, Lopes U, Mottram JC, Lima APCA. Neutrophil elastase promotes Leishmania donovani infection via interferon-β. FASEB J 2019; 33:10794-10807. [PMID: 31284755 PMCID: PMC6766642 DOI: 10.1096/fj.201900524r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Visceral leishmaniasis is a deadly illness caused by Leishmania donovani that provokes liver and spleen inflammation and tissue destruction. In cutaneous leishmaniasis, the protein of L. major, named inhibitor of serine peptidases (ISP) 2, inactivates neutrophil elastase (NE) present at the macrophage surface, resulting in blockade of TLR4 activation, prevention of TNF-α and IFN-β production, and parasite survival. We report poor intracellular growth of L. donovani in macrophages from knockout mice for NE (ela-/-), TLR4, or TLR2. NE and TLR4 colocalized with the parasite in the parasitophorous vacuole. Parasite load in the liver and spleen of ela-/- mice were reduced and accompanied by increased NO and decreased TGF-β production. Expression of ISP2 was not detected in L. donovani, and a transgenic line constitutively expressing ISP2, displayed poor intracellular growth in macrophages and decreased burden in mice. Infected ela-/- macrophages displayed significantly lower IFN-β mRNA than background mice macrophages, and the intracellular growth was fully restored by exogenous IFN-β. We propose that L. donovani utilizes the host NE-TLR machinery to induce IFN-β necessary for parasite survival and growth during early infection. Low or absent expression of parasite ISP2 in L. donovani is necessary to preserve the activation of the NE-TLR pathway.-Dias, B. T., Dias-Teixeira, K. L., Godinho, J. P., Faria, M. S., Calegari-Silva, T., Mukhtar, M. M., Lopes, U. G., Mottram, J. C., Lima, A. P. C. A. Neutrophil elastase promotes Leishmania donovani infection via interferon-β.
Collapse
Affiliation(s)
- Bruna T Dias
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Karina Luisa Dias-Teixeira
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Joseane P Godinho
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marilia S Faria
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Teresa Calegari-Silva
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maowia M Mukhtar
- Bioscience Research Institute, Ibn Sina University, Khartoum, Sudan
| | - Ulisses Lopes
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jeremy C Mottram
- Department of Biology, York Biomedical Research Institute, University of York, York, United Kingdom
| | - Ana Paula C A Lima
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
44
|
Wei F, Gong W, Wang J, Yang Y, Liu J, Wang Y, Cao J. Role of the lipoxin A4 receptor in the development of neutrophil extracellular traps in Leishmania infantum infection. Parasit Vectors 2019; 12:275. [PMID: 31142352 PMCID: PMC6542009 DOI: 10.1186/s13071-019-3530-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 05/23/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Neutrophils play an immunomodulatory role through the release of neutrophil extracellular traps (NETs). NETs are released in response to Leishmania infection, but the mechanism of NET extrusion has not been elucidated. The lipoxin A4 receptor on neutrophils is crucial for the inflammatory response and immune regulation of many diseases, including Leishmania infection. Therefore, in the present study, we tried to explore whether Leishmania infantum promastigotes stimulate neutrophil activation and NET release via activating the lipoxin A4 receptor. RESULTS Leishmania infantum promastigotes stimulated neutrophil activity, but blocking of the lipoxin A4 receptor with its antagonist Boc prior to L. infantum stimulation abrogated these effects. Neutrophils showed citrullinated histone H3 expression and simultaneous NET extrusion on L. infantum stimulation, but a decline in both was observed on blocking of the lipoxin A4 receptor. Moreover, differentiated HL-60 cells with lipoxin A4 receptor silencing showed a decrease in citrullinated histone H3 expression as compared to the unsilenced HL-60 samples on stimulation with promastigotes. CONCLUSIONS Leishmania infantum promastigotes altered the characteristics of neutrophils and induced NET extrusion by activating the lipoxin A4 receptor. The lipoxin A4 receptor may have potential as a therapeutic target in relation to NET extrusion in the treatment of leishmaniasis, but its mechanisms of action need to be explored in more depth.
Collapse
Affiliation(s)
- Furong Wei
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, China.,Chinese Center for Tropical Diseases Research, Shanghai, 200025, China.,WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, 200025, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025, China
| | - Wenci Gong
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, China.,Chinese Center for Tropical Diseases Research, Shanghai, 200025, China.,WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, 200025, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025, China
| | - Junyun Wang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, China.,Chinese Center for Tropical Diseases Research, Shanghai, 200025, China.,WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, 200025, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025, China
| | - Yuetao Yang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, China.,Chinese Center for Tropical Diseases Research, Shanghai, 200025, China.,WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, 200025, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025, China
| | - Jianxiu Liu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, China.,Chinese Center for Tropical Diseases Research, Shanghai, 200025, China.,WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, 200025, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025, China
| | - Yanjuan Wang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, China.,Chinese Center for Tropical Diseases Research, Shanghai, 200025, China.,WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, 200025, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025, China
| | - Jianping Cao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, China. .,Chinese Center for Tropical Diseases Research, Shanghai, 200025, China. .,WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, China. .,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, 200025, China. .,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025, China.
| |
Collapse
|
45
|
Polari LP, Carneiro PP, Macedo M, Machado PRL, Scott P, Carvalho EM, Bacellar O. Leishmania braziliensis Infection Enhances Toll-Like Receptors 2 and 4 Expression and Triggers TNF-α and IL-10 Production in Human Cutaneous Leishmaniasis. Front Cell Infect Microbiol 2019; 9:120. [PMID: 31119102 PMCID: PMC6507514 DOI: 10.3389/fcimb.2019.00120] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 04/04/2019] [Indexed: 12/14/2022] Open
Abstract
Cutaneous leishmaniasis (CL) caused by infection with Leishmania braziliensis is characterized by an exaggerated inflammatory response that controls the parasite burden, but also contributes to pathology. While myeloid cells are required to eliminate the parasite, recent studies indicate that they may also participate in the inflammatory response driving disease progression. The innate immune response to leishmania is driven in part by the Toll-like receptors (TLRs) TLR2, TLR4, and TLR9. In this study, we used flow cytometric analysis to compare TLR2 and TLR4 expression in monocyte subsets (classical, intermediate, and non-classical) from CL patients and healthy subjects (HS). We also determined if there was an association of either the pro-inflammatory cytokine TNF or the anti-inflammatory cytokine IL-10 with TLR2 or TLR4 expression levels after L. braziliensis infection. In vitro infection with L. braziliensis caused CL monocytes to up-regulate TLR2 and TLR4 expression. We also found that intermediate monocytes expressed the highest levels of TLR2 and TLR4 and that infected monocytes produced more TNF and IL-10 than uninfected monocytes. Finally, while classical and intermediate monocytes were mainly responsible for TNF production, classical monocytes were the main source of IL-10. Collectively, our studies revealed that up-regulated TLR2/4 expression and TNF production by intermediate/inflammatory subsets of monocytes from patients correlates with detrimental outcome of cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Ludmila P Polari
- Serviço de Imunologia, Complexo Hospitalar Universitário Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Pedro Paulo Carneiro
- Serviço de Imunologia, Complexo Hospitalar Universitário Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil.,Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais - INCT-DT (CNPq/MCT), Salvador, Brazil
| | - Michael Macedo
- Serviço de Imunologia, Complexo Hospitalar Universitário Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Paulo R L Machado
- Serviço de Imunologia, Complexo Hospitalar Universitário Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil.,Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais - INCT-DT (CNPq/MCT), Salvador, Brazil
| | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Edgar M Carvalho
- Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais - INCT-DT (CNPq/MCT), Salvador, Brazil.,Instituto Pesquisa Gonçalo Moniz - Fiocruz-Bahia, Salvador, Brazil
| | - Olívia Bacellar
- Serviço de Imunologia, Complexo Hospitalar Universitário Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil.,Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais - INCT-DT (CNPq/MCT), Salvador, Brazil
| |
Collapse
|
46
|
Rossi M, Fasel N. How to master the host immune system? Leishmania parasites have the solutions! Int Immunol 2019; 30:103-111. [PMID: 29294040 PMCID: PMC5892169 DOI: 10.1093/intimm/dxx075] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 12/21/2017] [Indexed: 12/11/2022] Open
Abstract
Infection by protozoan parasites of the genus Leishmania results in the development of leishmaniasis, an increasingly prevalent group of diseases affecting over 12 million people worldwide. Leishmaniasis can have very different outcomes ranging from cutaneous lesions, mucosal lesions to visceralization depending on the species of the infecting parasite and on the immune response developed by the host. As an obligate intracellular parasite, residing within macrophages, Leishmania evolved in strict contact with the host immune system, developing different mechanisms to evade or modulate the immune response. Various types of immune responses are observed during different Leishmania spp. infections, resulting in parasite clearance but also contributing to the pathogenesis, thus increasing the complexity of the course of the disease. Interestingly, depending on the type of leishmaniasis developed, opposite treatment strategies, which either boost or inhibit the inflammatory response, have shown efficacy. In this review, we summarize the contribution of different immune cell types to the development of the anti-leishmanial immune response and the parasite strategies to evade and modulate host immunity. Further, we discuss the involvement of co-infecting pathogens in the determination of the outcome of leishmaniasis and on the effectiveness of treatment and the implication of the immune response for treatment and vaccine development.
Collapse
Affiliation(s)
- Matteo Rossi
- Department of Biochemistry, University of Lausanne, Epalinges, Lausanne, Switzerland
| | - Nicolas Fasel
- Department of Biochemistry, University of Lausanne, Epalinges, Lausanne, Switzerland
| |
Collapse
|
47
|
Valério-Bolas A, Pereira M, Alexandre-Pires G, Santos-Mateus D, Rodrigues A, Rafael-Fernandes M, Gabriel A, Passero F, Santos-Gomes G. Intracellular and extracellular effector activity of mouse neutrophils in response to cutaneous and visceral Leishmania parasites. Cell Immunol 2019; 335:76-84. [DOI: 10.1016/j.cellimm.2018.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/06/2018] [Accepted: 11/06/2018] [Indexed: 02/04/2023]
|
48
|
Vicente CR, Falqueto A. Differentiation of mucosal lesions in mucocutaneous leishmaniasis and paracoccidioidomycosis. PLoS One 2018; 13:e0208208. [PMID: 30475920 PMCID: PMC6258372 DOI: 10.1371/journal.pone.0208208] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/13/2018] [Indexed: 11/23/2022] Open
Abstract
Mucocutaneous leishmaniasis and paracoccidioidomycosis are infectious diseases with similar epidemiological and clinical aspects. Cases of both diseases may manifest similar lesions in the mucosa. Therefore, the determination of distinguishing characteristics for the purpose of differential diagnosis is critical for better management of the diseases. The present study evaluated factors that assist in the differentiation of mucosal lesions between these diseases. This cross-sectional study included data from medical records of 122 cases of mucocutaneous leishmaniasis and 83 cases of paracoccidioidomycosis attended at the university hospital Cassiano Antonio Moraes, located in Vitória, Espírito Santo State, Brazil. Comparison between the diseases included the following variables: sex, age, time of disease evolution, location of the lesion and symptoms. Adults and males were affected by both diseases at higher rates. Lesions in the nasal region (95.1%; p-value = 0.000) and the pharynx (20.5%; p-value = 0.009) and nasal obstruction (34.4%; p-value = 0.000) were associated with leishmaniasis. Paracoccidioidomycosis was associated with lesions in the oral region (90.4%; p-value = 0.000), oral pain (16.9%; p-value = 0.000), and hoarseness (14.5%; p-value = 0.008). In leishmaniasis, lesions in oral regions were not associated with oral pain and were frequently located close to the nasal area. The manifestations cited above could improve the differential diagnosis of leishmaniasis and paracoccidioidomycosis, and thereby potentially aid in the choice of appropriate confirmatory diagnostic testing.
Collapse
Affiliation(s)
- Creuza Rachel Vicente
- Department of Social Medicine, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo State, Brazil
- * E-mail:
| | - Aloisio Falqueto
- Department of Social Medicine, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo State, Brazil
| |
Collapse
|
49
|
Thomazelli APFDS, Tomiotto-Pellissier F, Miranda-Sapla MM, da Silva SS, Alvarenga DS, Panis C, Cataneo AHD, Bordignon J, Silveira GF, Yamauchi LM, de Sá JPSR, Felipe I, Pavanelli WR, Conchon-Costa I. Concanavalin-A displays leishmanicidal activity by inducing ROS production in human peripheral blood mononuclear cells. Immunopharmacol Immunotoxicol 2018; 40:387-392. [DOI: 10.1080/08923973.2018.1510960] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
| | - Fernanda Tomiotto-Pellissier
- Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brasil
| | - Milena Menegazzo Miranda-Sapla
- Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brasil
| | - Suelen Santos da Silva
- Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brasil
| | - Daniele Sapede Alvarenga
- Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brasil
| | - Carolina Panis
- Laboratory of Inflammatory Mediators, University of Western Paraná, UNIOESTE, Francisco Beltrão, Paraná, Brasil
| | - Allan Henrique Depieri Cataneo
- Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brasil
| | | | | | - Lucy Megumi Yamauchi
- Department of Microbiology, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brasil
| | | | - Ionice Felipe
- Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brasil
| | - Wander Rogério Pavanelli
- Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brasil
| | - Ivete Conchon-Costa
- Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brasil
| |
Collapse
|
50
|
Ontoria E, Hernández-Santana YE, González-García AC, López MC, Valladares B, Carmelo E. Transcriptional Profiling of Immune-Related Genes in Leishmania infantum-Infected Mice: Identification of Potential Biomarkers of Infection and Progression of Disease. Front Cell Infect Microbiol 2018; 8:197. [PMID: 30013952 PMCID: PMC6036295 DOI: 10.3389/fcimb.2018.00197] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/28/2018] [Indexed: 12/27/2022] Open
Abstract
Leishmania spp. is a protozoan parasite that affects millions of people around the world. At present, there is no effective vaccine to prevent leishmaniases in humans. A major limitation in vaccine development is the lack of precise understanding of the particular immunological mechanisms that allow parasite survival in the host. The parasite-host cell interaction induces dramatic changes in transcriptome patterns in both organisms, therefore, a detailed analysis of gene expression in infected tissues will contribute to the evaluation of drug and vaccine candidates, the identification of potential biomarkers, and the understanding of the immunological pathways that lead to protection or progression of disease. In this large-scale analysis, differential expression of 112 immune-related genes has been analyzed using high-throughput qPCR in spleens of infected and naïve Balb/c mice at four different time points. This analysis revealed that early response against Leishmania infection is characterized by the upregulation of Th1 markers and M1-macrophage activation molecules such as Ifng, Stat1, Cxcl9, Cxcl10, Ccr5, Cxcr3, Xcl1, and Ccl3. This activation doesn't protect spleen from infection, since parasitic burden rises along time. This marked difference in gene expression between infected and control mice disappears during intermediate stages of infection, probably related to the strong anti-inflammatory and immunosuppresory signals that are activated early upon infection (Ctla4) or remain activated throughout the experiment (Il18bp). The overexpression of these Th1/M1 markers is restored later in the chronic phase (8 wpi), suggesting the generation of a classical "protective response" against leishmaniasis. Nonetheless, the parasitic burden rockets at this timepoint. This apparent contradiction can be explained by the generation of a regulatory immune response characterized by overexpression of Ifng, Tnfa, Il10, and downregulation Il4 that counteracts the Th1/M1 response. This large pool of data was also used to identify potential biomarkers of infection and parasitic burden in spleen, on the bases of two different regression models. Given the results, gene expression signature analysis appears as a useful tool to identify mechanisms involved in disease outcome and to establish a rational approach for the identification of potential biomarkers useful for monitoring disease progression, new therapies or vaccine development.
Collapse
Affiliation(s)
- Eduardo Ontoria
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, La Laguna, Spain
| | - Yasmina E. Hernández-Santana
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, La Laguna, Spain
| | - Ana C. González-García
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, La Laguna, Spain
| | - Manuel C. López
- Departamento de Biología Molecular, Instituto de Parasitología y Biomedicina “López Neyra”, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Basilio Valladares
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, La Laguna, Spain
| | - Emma Carmelo
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, La Laguna, Spain
| |
Collapse
|