1
|
Palomino-Cano C, Moreno E, Irache JM, Espuelas S. Targeting and activation of macrophages in leishmaniasis. A focus on iron oxide nanoparticles. Front Immunol 2024; 15:1437430. [PMID: 39211053 PMCID: PMC11357945 DOI: 10.3389/fimmu.2024.1437430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Macrophages play a pivotal role as host cells for Leishmania parasites, displaying a notable functional adaptability ranging from the proinflammatory, leishmanicidal M1 phenotype to the anti-inflammatory, parasite-permissive M2 phenotype. While macrophages can potentially eradicate amastigotes through appropriate activation, Leishmania employs diverse strategies to thwart this activation and redirect macrophages toward an M2 phenotype, facilitating its survival and replication. Additionally, a competition for iron between the two entities exits, as iron is vital for both and is also implicated in macrophage defensive oxidative mechanisms and modulation of their phenotype. This review explores the intricate interplay between macrophages, Leishmania, and iron. We focus the attention on the potential of iron oxide nanoparticles (IONPs) as a sort of immunotherapy to treat some leishmaniasis forms by reprogramming Leishmania-permissive M2 macrophages into antimicrobial M1 macrophages. Through the specific targeting of iron in macrophages, the use of IONPs emerges as a promising strategy to finely tune the parasite-host interaction, endowing macrophages with an augmented antimicrobial arsenal capable of efficiently eliminating these intrusive microbes.
Collapse
Affiliation(s)
- Carmen Palomino-Cano
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Esther Moreno
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Juan M. Irache
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Medical Research Institute (IdiSNA), Pamplona, Spain
| | - Socorro Espuelas
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Medical Research Institute (IdiSNA), Pamplona, Spain
| |
Collapse
|
2
|
Reyaz E, Puri N, Selvapandiyan A. Global Remodeling of Host Proteome in Response to Leishmania Infection. ACS Infect Dis 2024; 10:5-19. [PMID: 38084821 DOI: 10.1021/acsinfecdis.3c00492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
The protozoan parasite Leishmania possesses an intrinsic ability to modulate a multitude of pathways in the host, toward aiding its own proliferation. In response, the host reprograms its cellular, immunological, and metabolic machinery to evade the parasite's lethal impact. Besides inducing various antioxidant signaling pathways to counter the elevated stress response proteins like heme oxygenase-1 (HO-1), Leishmania also attempts to delay host cell apoptosis by promoting anti-apoptotic proteins like Bcl-2. The downstream modulation of apoptotic proteins is regulated by effector pathways, including the PI3K/Akt survival pathway, the mitogen-activated protein kinases (MAPKs) signaling pathway, and STAT phosphorylation. In addition, Leishmania assists in its infection in a time-dependent manner by modulating the level of various proteins of autophagic machinery. Immune effector cells, such as mast cells and neutrophils, entrap and kill the pathogen by secreting various granular proteins. In contrast, the host macrophages exert their leishmanicidal effect by secreting various cytokines, such as IL-2, IL-12, etc. An interplay of various signaling pathways occurs in an organized network that is highly specific to both pathogen and host species. This Review analyzes the modulation of expression of proteins, including the cytokines, providing a realistic approach toward understanding the pathophysiology of disease and predicting some prominent markers for disease intervention and vaccine support strategies.
Collapse
Affiliation(s)
- Enam Reyaz
- Department of Molecular Medicine, Jamia Hamdard, New Delhi 110062, India
| | - Niti Puri
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | | |
Collapse
|
3
|
Kot K, Kupnicka P, Tarnowski M, Tomasiak P, Kosik-Bogacka D, Łanocha-Arendarczyk N. The role of apoptosis and oxidative stress in the pathophysiology of Acanthamoeba spp. infection in the kidneys of hosts with different immunological status. Parasit Vectors 2023; 16:445. [PMID: 38041167 PMCID: PMC10693070 DOI: 10.1186/s13071-023-06052-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/10/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND Acanthamoeba spp. are opportunistic pathogens that cause inflammation, mostly in the brain, lungs and cornea. Recent reports indicate kidney dysfunction in hosts with systemic acanthamoebiasis. The aim of the study was to analyze the gene expression and protein concentration of NADPH oxidase 2 and 4 (NOX2 and NOX4, respectively) and nuclear erythroid 2-related factor (Nrf2) in the kidneys of hosts with systemic acanthamoebiasis. We also aimed to determine the protein and gene expressions of Bcl2, Bax, caspases 3 and 9. METHODS Mice were divided into four groups based on their immunological status and Acanthamoeba sp. infection: A, immunocompetent Acanthamoeba sp.-infected mice; AS, immunosuppressed Acanthamoeba sp.- infected mice; C, immunocompetent uninfected mice; CS, immunosuppressed uninfected mice. NOX2, NOX4 and Nrf2 were analyzed by quantitative reverse transcription PCR (qRT-PCR) and ELISA methods, while pro-apoptotic and anti-apoptotic proteins (Bax and Bcl-2, respectively), Cas9, Cas3 were analyzed by qRT-PCR and western blot methods. RESULTS: Increased gene expression and/or protein concentration of NOX2 and NOX4 were found in both immunocompetent and immunosuppressed mice infected with Acanthamoeba sp. (groups A and AS, respectively). Gene expression and/or protein concentration of Nrf2 were higher in group A than in control animals. Compared to control mice, in the AS group the expression of the Nrf2 gene was upregulated while the concentration of Nrf2 protein was decreased. Additionally in A group, higher gene and protein expression of Bcl-2, and lower gene as well as protein expression of Bax, caspases 3 and 9 were noted. In contrast, the AS group showed lower gene and protein expression of Bcl-2, and higher gene as well as protein expression of Bax, caspases 3 and 9. CONCLUSIONS This study is the first to address the mechanisms occurring in the kidneys of hosts infected with Acanthamoeba sp. The contact of Acanthamoeba sp. with the host cell surface and/or the oxidative burst caused by elevated levels of NOXs lead to an antioxidant response enhanced by the Nrf2 pathway. Acanthamoeba sp. have various strategies concerning apoptosis. In immunocompetent hosts, amoebae inhibit the apoptosis of kidney cells, and in immunosuppressed hosts, they lead to increased apoptosis by the intrinsic pathway and thus to a more severe course of the disease.
Collapse
Affiliation(s)
- Karolina Kot
- Department of Biology and Medical Parasitology, Pomeranian Medical University in Szczecin, Szczecin, Poland.
| | - Patrycja Kupnicka
- Department of Biochemistry, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Maciej Tarnowski
- Department of Physiology in Health Sciences, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Patrycja Tomasiak
- Institute of Physical Culture Sciences, University of Szczecin, Szczecin, Poland
| | - Danuta Kosik-Bogacka
- Independent Laboratory of Pharmaceutical Botany, Department of Biology and Medical Parasitology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | | |
Collapse
|
4
|
Anuntasomboon P, Siripattanapipong S, Unajak S, Choowongkomon K, Burchmore R, Leelayoova S, Mungthin M, E-Kobon T. Identification of a unique conserved region from a kinetoplastid genome of Leishmania orientalis (formerly named Leishmania siamensis) strain PCM2 in Thailand. Sci Rep 2023; 13:19644. [PMID: 37950023 PMCID: PMC10638283 DOI: 10.1038/s41598-023-46638-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023] Open
Abstract
Mitochondrial DNAs (mtDNAs) appear in almost all eukaryotic species and are useful molecular markers for phylogenetic studies and species identification. Kinetoplast DNAs (kDNAs) are structurally complex circular mtDNA networks in kinetoplastids, divided into maxicircles and minicircles. Despite several kDNAs of many Leishmania species being examined, the kDNAs of the new species, Leishmania orientalis (formerly named Leishmania siamensis) strain PCM2, have not been explored. This study aimed to investigate the maxicircle and minicircle DNAs of L. orientalis strain PCM2 using hybrid genome sequencing technologies and bioinformatic analyses. The kDNA sequences were isolated and assembled using the SPAdes hybrid assembler from the Illumina short-read and PacBio long-read data. Circular contigs of the maxicircle and minicircle DNAs were reconstructed and confirmed by BLASTn and rKOMICs programs. The kDNA genome was annotated by BLASTn before the genome comparison and phylogenetic analysis by progressiveMauve, MAFFT, and MEGA programs. The maxicircle of L. orientalis strain PCM2 (18,215 bp) showed 99.92% similarity and gene arrangement to Leishmania enriettii strain LEM3045 maxicircle with variation in the 12s rRNA gene and divergent region. Phylogenetics of the whole sequence, coding regions, divergent regions, and 12s rRNA gene also confirmed this relationship and subgenera separation. The identified 105 classes of minicircles (402-1177 bp) were clustered monophyletically and related to the Leishmania donovani minicircles. The kinetoplast maxicircle and minicircle DNAs of L. orientalis strain PCM2 contained a unique conserved region potentially useful for specific diagnosis of L. orientalis and further exploration of this parasite population genetics in Thailand and related regions.
Collapse
Affiliation(s)
- Pornchai Anuntasomboon
- Department of Genetics, Faculty of Science, Kasetsart University, Bangkok, Thailand
- Omics Center for Agriculture, Bioresources, Food, and Health, Kasetsart University (OmiKU), Bangkok, Thailand
| | | | - Sasimanas Unajak
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | | | - Richard Burchmore
- Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Saovanee Leelayoova
- Department of Parasitology, Phramongkutklao College of Medicine, Bangkok, Thailand
| | - Mathirut Mungthin
- Department of Parasitology, Phramongkutklao College of Medicine, Bangkok, Thailand
| | - Teerasak E-Kobon
- Department of Genetics, Faculty of Science, Kasetsart University, Bangkok, Thailand.
- Omics Center for Agriculture, Bioresources, Food, and Health, Kasetsart University (OmiKU), Bangkok, Thailand.
| |
Collapse
|
5
|
Pacheco-Fernandez T, Markle H, Verma C, Huston R, Gannavaram S, Nakhasi HL, Satoskar AR. Field-Deployable Treatments For Leishmaniasis: Intrinsic Challenges, Recent Developments and Next Steps. Res Rep Trop Med 2023; 14:61-85. [PMID: 37492219 PMCID: PMC10364832 DOI: 10.2147/rrtm.s392606] [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: 03/01/2023] [Accepted: 06/08/2023] [Indexed: 07/27/2023] Open
Abstract
Leishmaniasis is a neglected tropical disease endemic primarily to low- and middle-income countries, for which there has been inadequate development of affordable, safe, and efficacious therapies. Clinical manifestations of leishmaniasis range from self-healing skin lesions to lethal visceral infection with chances of relapse. Although treatments are available, secondary effects limit their use outside the clinic and negatively impact the quality of life of patients in endemic areas. Other non-medicinal treatments, such as thermotherapies, are limited to use in patients with cutaneous leishmaniasis but not with visceral infection. Recent studies shed light to mechanisms through which Leishmania can persist by hiding in cellular safe havens, even after chemotherapies. This review focuses on exploring the cellular niches that Leishmania parasites may be leveraging to persist within the host. Also, the cellular, metabolic, and molecular implications of Leishmania infection and how those could be targeted for therapeutic purposes are discussed. Other therapies, such as those developed against cancer or for manipulation of the ferroptosis pathway, are proposed as possible treatments against leishmaniasis due to their mechanisms of action. In particular, treatments that target hematopoietic stem cells and monocytes, which have recently been found to be necessary components to sustain the infection and provide a safe niche for the parasites are discussed in this review as potential field-deployable treatments against leishmaniasis.
Collapse
Affiliation(s)
- Thalia Pacheco-Fernandez
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Hannah Markle
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Chaitenya Verma
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43201, USA
| | - Ryan Huston
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43201, USA
- Department of Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43201, USA
| | - Sreenivas Gannavaram
- Division of Emerging and Transfusion Transmitted Disease, 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
| | - Abhay R Satoskar
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43201, USA
- Department of Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43201, USA
| |
Collapse
|
6
|
Moreno CJG, Farias HM, de Lima Medeiros R, de Brito Pinto TK, de Freitas Oliveira JW, de Sousa FL, de Medeiros MJC, Amorim-Carmo B, Santos-Gomes G, de Lima Pontes D, Rocha HAO, Frazão NF, Silva MS. Quantum Biochemistry Screening and In Vitro Evaluation of Leishmania Metalloproteinase Inhibitors. Int J Mol Sci 2022; 23:8553. [PMID: 35955687 PMCID: PMC9368959 DOI: 10.3390/ijms23158553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/16/2022] [Accepted: 07/17/2022] [Indexed: 12/04/2022] Open
Abstract
Leishmanolysin, also known as major promastigote protease (PSP) or gp63, is the most abundant surface glycoprotein of Leishmania spp., and has been extensively studied and recognized as the main parasite virulence factor. Characterized as a metalloprotease, gp63 can be powerfully inactivated in the presence of a metal chelator. In this study, we first used the structural parameters of a 7-hydroxycoumarin derivative, L1 compound, to evaluate the theoretical-computational experiments against gp63, comparing it with an available metal chelator already described. The methodology followed was (i) analysis of the three-dimensional structure of gp63 as well as its active site, and searching the literature and molecular databases for possible inhibitors; (ii) molecular docking simulations and investigation of the interactions in the generated protein-ligand complexes; and (iii) the individual energy of the gp63 amino acids that interacted most with the ligands of interest was quantified by ab initio calculations using Molecular Fraction with Conjugated Caps (MFCC). MFCC still allowed the final quantum balance calculations of the protein interaction to be obtained with each inhibitor candidate binder. L1 obtained the best energy quantum balance result with -2 eV, followed by DETC (-1.4 eV), doxycycline (-1.3 eV), and 4-terpineol (-0.6 eV), and showed evidence of covalent binding in the enzyme active site. In vitro experiments confirmed L1 as highly effective against L. amazonensis parasites. The compound also exhibited a low cytotoxicity profile against mammalian RAW and 3T3 cells lines, presenting a selective index of 149.19 and 380.64 µM, respectively. L1 induced promastigote forms' death by necrosis and the ultrastructural analysis revealed disruption in membrane integrity. Furthermore, leakage of the contents and destruction of the parasite were confirmed by Spectroscopy Dispersion analysis. These results together suggested L1 has a potential effect against L. amazonensis, the etiologic agent of diffuse leishmaniasis, and the only one that currently does not have a satisfactory treatment.
Collapse
Affiliation(s)
- Cláudia Jassica Gonçalves Moreno
- Laboratory of Immunoparasitology, Department of Clinical and Toxicological Analysis, Health Sciences Center, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil; (C.J.G.M.); (J.W.d.F.O.)
- Postgraduate Program in Pharmaceutical Sciences, Health Sciences Center, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil;
- Postgraduate Program in Biochemistry, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil;
| | - Henriqueta Monalisa Farias
- Academic Unit of Physics, Mathematics of the Education and Health Center, Federal University of Campina Grande, Campina Grande 58428-830, Brazil; (H.M.F.); (R.d.L.M.); (N.F.F.)
| | - Rafael de Lima Medeiros
- Academic Unit of Physics, Mathematics of the Education and Health Center, Federal University of Campina Grande, Campina Grande 58428-830, Brazil; (H.M.F.); (R.d.L.M.); (N.F.F.)
| | - Talita Katiane de Brito Pinto
- Postgraduate Program in Health Sciences, Health Sciences Center, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil;
| | - Johny Wysllas de Freitas Oliveira
- Laboratory of Immunoparasitology, Department of Clinical and Toxicological Analysis, Health Sciences Center, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil; (C.J.G.M.); (J.W.d.F.O.)
- Postgraduate Program in Biochemistry, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil;
| | - Francimar Lopes de Sousa
- Laboratory of Chemistry of Coordination and Polymers (LQCPol), Institute of Chemistry Chemistry Institute, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil; (F.L.d.S.J.); (M.J.C.d.M.); (D.d.L.P.)
| | - Mayara Jane Campos de Medeiros
- Laboratory of Chemistry of Coordination and Polymers (LQCPol), Institute of Chemistry Chemistry Institute, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil; (F.L.d.S.J.); (M.J.C.d.M.); (D.d.L.P.)
| | - Bruno Amorim-Carmo
- Postgraduate Program in Pharmaceutical Sciences, Health Sciences Center, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil;
| | - Gabriela Santos-Gomes
- Global Health and Tropical Medicine, GHTM, Institute of Hygiene and Tropical Medicine, IHMT, NOVA University of Lisbon—UNL, 1349-008 Lisbon, Portugal;
| | - Daniel de Lima Pontes
- Laboratory of Chemistry of Coordination and Polymers (LQCPol), Institute of Chemistry Chemistry Institute, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil; (F.L.d.S.J.); (M.J.C.d.M.); (D.d.L.P.)
| | - Hugo Alexandre Oliveira Rocha
- Postgraduate Program in Biochemistry, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil;
- Postgraduate Program in Health Sciences, Health Sciences Center, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil;
| | - Nilton Fereira Frazão
- Academic Unit of Physics, Mathematics of the Education and Health Center, Federal University of Campina Grande, Campina Grande 58428-830, Brazil; (H.M.F.); (R.d.L.M.); (N.F.F.)
| | - Marcelo Sousa Silva
- Laboratory of Immunoparasitology, Department of Clinical and Toxicological Analysis, Health Sciences Center, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil; (C.J.G.M.); (J.W.d.F.O.)
- Postgraduate Program in Pharmaceutical Sciences, Health Sciences Center, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil;
- Postgraduate Program in Biochemistry, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil;
- Global Health and Tropical Medicine, GHTM, Institute of Hygiene and Tropical Medicine, IHMT, NOVA University of Lisbon—UNL, 1349-008 Lisbon, Portugal;
| |
Collapse
|
7
|
Chakrabarti A, Narayana C, Joshi N, Garg S, Garg LC, Ranganathan A, Sagar R, Pati S, Singh S. Metalloprotease Gp63-Targeting Novel Glycoside Exhibits Potential Antileishmanial Activity. Front Cell Infect Microbiol 2022; 12:803048. [PMID: 35601095 PMCID: PMC9115111 DOI: 10.3389/fcimb.2022.803048] [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: 10/27/2021] [Accepted: 03/14/2022] [Indexed: 11/17/2022] Open
Abstract
Visceral leishmaniasis (VL) and post kala-azar dermal leishmaniasis (PKDL) affect most of the poor populations worldwide. The current treatment modalities include liposomal formulation or deoxycholate salt of amphotericin B, which has been associated with various complications and severe side effects. Encouraged from the recent marked antimalarial effects from plant-derived glycosides, in this study, we have exploited a green chemistry-based approach to chemically synthesize a library of diverse glycoside derivatives (Gly1–12) and evaluated their inhibitory efficacy against the AG83 strain of Leishmania donovani. Among the synthesized glycosides, the in vitro inhibitory activity of Glycoside-2 (Gly2) (1.13 µM IC50 value) on L. donovani promastigote demonstrated maximum cytotoxicity with ~94% promastigote death as compared to amphotericin B that was taken as a positive control. The antiproliferative effect of Gly2 on promastigote encouraged us to analyze the structure–activity relationship of Gly2 with Gp63, a zinc metalloprotease that majorly localizes at the surface of the promastigote and has a role in its development and multiplication. The result demonstrated the exceptional binding affinity of Gly2 toward the catalytic domain of Gp63. These data were thereafter validated through cellular thermal shift assay in a physiologically relevant cellular environment. Mechanistically, reduced multiplication of promastigotes on treatment with Gly2 induces the destabilization of redox homeostasis in promastigotes by enhancing reactive oxygen species (ROS), coupled with depolarization of the mitochondrial membrane. Additionally, Gly2 displayed strong lethal effects on infectivity and multiplication of amastigote inside the macrophage in the amastigote–macrophage infection model in vitro as compared to amphotericin B treatment. Gp63 is also known to bestow protection against complement-mediated lysis of parasites. Interestingly, Gly2 treatment enhances the complement-mediated lysis of L. donovani promastigotes in serum physiological conditions. In addition, Gly2 was found to be equally effective against the clinical promastigote forms of PKDL strain (IC50 value of 1.97 µM); hence, it could target both VL and PKDL simultaneously. Taken together, this study reports the serendipitous discovery of Gly2 with potent antileishmanial activity and proves to be a novel chemotherapeutic prototype against VL and PKDL.
Collapse
Affiliation(s)
- Amrita Chakrabarti
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, India
| | - Chintam Narayana
- Department of Chemistry, School of Natural Sciences, Shiv Nadar University, Greater Noida, India
| | - Nishant Joshi
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, India
| | - Swati Garg
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, India
- Special Centre for Molecular Medicine, Jawaharlal Nehru University (JNU), New Delhi, India
| | - Lalit C. Garg
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi, India
| | - Anand Ranganathan
- Special Centre for Molecular Medicine, Jawaharlal Nehru University (JNU), New Delhi, India
| | - Ram Sagar
- Department of Chemistry, School of Natural Sciences, Shiv Nadar University, Greater Noida, India
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, India
- *Correspondence: Shailja Singh, ; Soumya Pati, ; Ram Sagar,
| | - Soumya Pati
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, India
- *Correspondence: Shailja Singh, ; Soumya Pati, ; Ram Sagar,
| | - Shailja Singh
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, India
- Special Centre for Molecular Medicine, Jawaharlal Nehru University (JNU), New Delhi, India
- *Correspondence: Shailja Singh, ; Soumya Pati, ; Ram Sagar,
| |
Collapse
|
8
|
Cecílio P, Cordeiro-da-Silva A, Oliveira F. Sand flies: Basic information on the vectors of leishmaniasis and their interactions with Leishmania parasites. Commun Biol 2022; 5:305. [PMID: 35379881 PMCID: PMC8979968 DOI: 10.1038/s42003-022-03240-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/08/2022] [Indexed: 11/09/2022] Open
Abstract
Blood-sucking arthropods transmit a variety of human pathogens acting as disseminators of the so-called vector-borne diseases. Leishmaniasis is a spectrum of diseases caused by different Leishmania species, transmitted quasi worldwide by sand flies. However, whereas many laboratories focus on the disease(s) and etiological agents, considerably less study the respective vectors. In fact, information on sand flies is neither abundant nor easy to find; aspects including basic biology, ecology, and sand-fly-Leishmania interactions are usually reported separately. Here, we compile elemental information on sand flies, in the context of leishmaniasis. We discuss the biology, distribution, and life cycle, the blood-feeding process, and the Leishmania-sand fly interactions that govern parasite transmission. Additionally, we highlight some outstanding questions that need to be answered for the complete understanding of parasite–vector–host interactions in leishmaniasis. In this review, numerous aspects of sand flies as vectors of Leishmania parasites—from biology to the vector parasite interactions—are discussed.
Collapse
Affiliation(s)
- Pedro Cecílio
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA. .,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal. .,Parasite Disease Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal. .,Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto (FFUP), Porto, Portugal.
| | - Anabela Cordeiro-da-Silva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Parasite Disease Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto (FFUP), Porto, Portugal
| | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
| |
Collapse
|
9
|
Kolářová I, Valigurová A. Hide-and-Seek: A Game Played between Parasitic Protists and Their Hosts. Microorganisms 2021; 9:2434. [PMID: 34946036 PMCID: PMC8707157 DOI: 10.3390/microorganisms9122434] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/17/2021] [Accepted: 11/20/2021] [Indexed: 11/17/2022] Open
Abstract
After invading the host organism, a battle occurs between the parasitic protists and the host's immune system, the result of which determines not only whether and how well the host survives and recovers, but also the fate of the parasite itself. The exact weaponry of this battle depends, among others, on the parasite localisation. While some parasitic protists do not invade the host cell at all (extracellular parasites), others have developed successful intracellular lifestyles (intracellular parasites) or attack only the surface of the host cell (epicellular parasites). Epicellular and intracellular protist parasites have developed various mechanisms to hijack host cell functions to escape cellular defences and immune responses, and, finally, to gain access to host nutrients. They use various evasion tactics to secure the tight contact with the host cell and the direct nutrient supply. This review focuses on the adaptations and evasion strategies of parasitic protists on the example of two very successful parasites of medical significance, Cryptosporidium and Leishmania, while discussing different localisation (epicellular vs. intracellular) with respect to the host cell.
Collapse
Affiliation(s)
- Iva Kolářová
- Laboratory of Vector Biology, Department of Parasitology, Faculty of Science, Charles University, Albertov 6, 128 44 Prague, Czech Republic
| | - Andrea Valigurová
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic
| |
Collapse
|
10
|
Apuzzo CF, Sullivan EC, Platt DC, Seger-Held I, Jones MA. Leishmania tarentolae novel responses to Bi 3+-doped strontium aluminum oxyfluorides. Heliyon 2021; 7:e07896. [PMID: 34504976 PMCID: PMC8414179 DOI: 10.1016/j.heliyon.2021.e07896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 08/19/2021] [Accepted: 08/27/2021] [Indexed: 11/20/2022] Open
Abstract
Novel therapeutics for the treatment of leishmaniasis are of interest as the disease not only is becoming more prevalent, but drug resistance is increasing in certain regions of the world. Reported here is the use of Bi3+-doped strontium aluminum oxyfluoride phosphors and protease inhibitors to test in vitro inhibitory activity against cultured promastigote Leishmania tarentolae and effects on L. tarentolae secreted acid phosphatase (SAP) activity. Cell viability did not significantly decrease in the presence of 50 μM anti-perovskite compounds, implying limited cytotoxicity. Yet SAP activity did increase in the cell free preparations with time in the presence of strontium compounds. Of interest was the observation that cell free SAP activity did not increase in the presence of protease inhibitors with or without added strontium compounds. Since secreted proteases may play a role in the maturation of Leishmania SAP and thus be involved with parasite-host infection establishment, this is in further need of evaluation. Nitric oxide production on day 4 post-addition of the strontium compounds was evaluated and showed an approximately 50% decrease in NO production in the presence of two test compounds relative to DMSO control cells. This is the first report of anti-perovskite compound inhibition of NO production by Leishmania.
Collapse
Affiliation(s)
- C. Fiore Apuzzo
- Department of Chemistry, Campus Box 4160, Illinois State University, Normal, IL, 61790, USA
| | - Eirin C. Sullivan
- Department of Chemistry, University of North Florida, Jacksonville, FL, 32224, USA
| | - David C. Platt
- Department of Chemistry, Campus Box 4160, Illinois State University, Normal, IL, 61790, USA
| | - Ian Seger-Held
- Department of Chemistry, Campus Box 4160, Illinois State University, Normal, IL, 61790, USA
| | - Marjorie A. Jones
- Department of Chemistry, Campus Box 4160, Illinois State University, Normal, IL, 61790, USA
| |
Collapse
|
11
|
Sandfly Fever Sicilian Virus-Leishmania major co-infection modulates innate inflammatory response favoring myeloid cell infections and skin hyperinflammation. PLoS Negl Trop Dis 2021; 15:e0009638. [PMID: 34310619 PMCID: PMC8341699 DOI: 10.1371/journal.pntd.0009638] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/05/2021] [Accepted: 07/09/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The leishmaniases are a group of sandfly-transmitted diseases caused by species of the protozoan parasite, Leishmania. With an annual incidence of 1 million cases, 1 billion people living in Leishmania-endemic regions, and nearly 30,000 deaths each year, leishmaniasis is a major global public health concern. While phlebotomine sandflies are well-known as vectors of Leishmania, they are also the vectors of various phleboviruses, including Sandfly Fever Sicilian Virus (SFSV). Cutaneous leishmaniasis (CL), caused by Leishmania major (L. major), among other species, results in development of skin lesions on the infected host. Importantly, there exists much variation in the clinical manifestation between individuals. We propose that phleboviruses, vectored by and found in the same sandfly guts as Leishmania, may be a factor in determining CL severity. It was reported by our group that Leishmania exosomes are released into the gut of the sandfly vector and co-inoculated during blood meals, where they exacerbate CL skin lesions. We hypothesized that, when taking a blood meal, the sandfly vector infects the host with Leishmania parasites and exosomes as well as phleboviruses, and that this viral co-infection results in a modulation of leishmaniasis. METHODOLOGY/PRINCIPAL FINDINGS In vitro, we observed modulation by SFSV in MAP kinase signaling as well as in the IRF3 pathway that resulted in a pro-inflammatory phenotype. Additionally, we found that SFSV and L. major co-infection resulted in an exacerbation of leishmaniasis in vivo, and by using endosomal (Toll-like receptor) TLR3, and MAVS knock-out mice, deduced that SFSV's hyperinflammatory effect was TLR3- and MAVS-dependent. Critically, we observed that L. major and SFSV co-infected C57BL/6 mice demonstrated significantly higher parasite burden than mice solely infected with L. major. Furthermore, viral presence increased leukocyte influx in vivo. This influx was accompanied by elevated total extracellular vesicle numbers. Interestingly, L. major displayed higher infectiveness with coincident phleboviral infection compared to L. major infection alone. CONCLUSION/SIGNIFICANCE Overall our work represents novel findings that contribute towards understanding the causal mechanisms governing cutaneous leishmaniasis pathology. Better comprehension of the potential role of viral co-infection could lead to treatment regimens with enhanced effectiveness.
Collapse
|
12
|
Samant M, Sahu U, Pandey SC, Khare P. Role of Cytokines in Experimental and Human Visceral Leishmaniasis. Front Cell Infect Microbiol 2021; 11:624009. [PMID: 33680991 PMCID: PMC7930837 DOI: 10.3389/fcimb.2021.624009] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/22/2021] [Indexed: 12/16/2022] Open
Abstract
Visceral Leishmaniasis (VL) is the most fatal form of disease leishmaniasis. To date, there are no effective prophylactic measures and therapeutics available against VL. Recently, new immunotherapy-based approaches have been established for the management of VL. Cytokines, which are predominantly produced by helper T cells (Th) and macrophages, have received great attention that could be an effective immunotherapeutic approach for the treatment of human VL. Cytokines play a key role in forming the host immune response and in managing the formation of protective and non-protective immunities during infection. Furthermore, immune response mediated through different cytokines varies from different host or animal models. Various cytokines viz. IFN-γ, IL-2, IL-12, and TNF-α play an important role during protection, while some other cytokines viz. IL-10, IL-6, IL-17, TGF-β, and others are associated with disease progression. Therefore, comprehensive knowledge of cytokine response and their interaction with various immune cells is very crucial to determine appropriate immunotherapies for VL. Here, we have discussed the role of cytokines involved in VL disease progression or host protection in different animal models and humans that will determine the clinical outcome of VL and open the path for the development of rapid and accurate diagnostic tools as well as therapeutic interventions against VL.
Collapse
Affiliation(s)
- Mukesh Samant
- Cell and Molecular Biology Laboratory, Department of Zoology, Kumaun University, Almora, India
| | - Utkarsha Sahu
- Department of Microbiology, All India Institute of Medical Sciences, Bhopal, India
| | - Satish Chandra Pandey
- Cell and Molecular Biology Laboratory, Department of Zoology, Kumaun University, Almora, India
| | - Prashant Khare
- Department of Microbiology, All India Institute of Medical Sciences, Bhopal, India
| |
Collapse
|
13
|
Torres Suarez E, Granados-Falla DS, Robledo SM, Murillo J, Upegui Y, Delgado G. Antileishmanial activity of synthetic analogs of the naturally occurring quinolone alkaloid N-methyl-8-methoxyflindersin. PLoS One 2020; 15:e0243392. [PMID: 33370295 PMCID: PMC7769561 DOI: 10.1371/journal.pone.0243392] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 11/19/2020] [Indexed: 11/19/2022] Open
Abstract
Leishmaniasis is a neglected, parasitic tropical disease caused by an intracellular protozoan from the genus Leishmania. Quinoline alkaloids, secondary metabolites found in plants from the Rutaceae family, have antiparasitic activity against Leishmania sp. N-methyl-8-methoxyflindersin (1), isolated from the leaves of Raputia heptaphylla and also known as 7-methoxy-2,2-dimethyl-2H,5H,6H-pyran[3,2-c]quinolin-5-one, shows antiparasitic activity against Leishmania promastigotes and amastigotes. This study used in silico tools to identify synthetic quinoline alkaloids having structure similar to that of compound 1 and then tested these quinoline alkaloids for their in vitro antiparasitic activity against Leishmania (Viannia) panamensis, in vivo therapeutic response in hamsters suffering from experimental cutaneous leishmaniasis (CL), and ex vivo immunomodulatory potential in healthy donors' human peripheral blood (monocyte)-derived macrophages (hMDMs). Compounds 1 (natural), 2 (synthetic), and 8 (synthetic) were effective against intracellular promastigotes (9.9, 3.4, and 1.6 μg/mL medial effective concentration [EC50], respectively) and amastigotes (5.07, 7.94, and 1.91 μg/mL EC50, respectively). Compound 1 increased nitric oxide production in infected hMDMs and triggered necrosis-related ultrastructural alterations in intracellular amastigotes, while compound 2 stimulated oxidative breakdown in hMDMs and caused ultrastructural alterations in the parasite 4 h posttreatment, and compound 8 failed to induce macrophage modulation but selectively induced apoptosis of infected hMDMs and alterations in the intracellular parasite ultrastructure. In addition, synthetic compounds 2 and 8 improved the health of hamsters suffering from experimental CL, without evidence of treatment-associated adverse toxic effects. Therefore, synthetic compounds 2 and 8 are potential therapeutic candidates for topical treatment of CL.
Collapse
Affiliation(s)
- Elaine Torres Suarez
- Grupo de Investigación en Inmunotoxicología, Departamento de Farmacia, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Diana Susana Granados-Falla
- Grupo de Investigación en Inmunotoxicología, Departamento de Farmacia, Universidad Nacional de Colombia, Bogotá, Colombia
- Vicerrectoría de Investigaciones, Universidad El Bosque, Bogotá, Colombia
| | - Sara María Robledo
- PECET, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Javier Murillo
- PECET, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Yulieth Upegui
- PECET, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Gabriela Delgado
- Grupo de Investigación en Inmunotoxicología, Departamento de Farmacia, Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
14
|
Ferreira-Paes T, Charret KDS, Ribeiro MRDS, Rodrigues RF, Leon LL. Comparative analysis of biological aspects of Leishmania infantum strains. PLoS One 2020; 15:e0230545. [PMID: 33270636 PMCID: PMC7714135 DOI: 10.1371/journal.pone.0230545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 11/13/2020] [Indexed: 11/19/2022] Open
Abstract
Leishmania infantum infantum (LII) is one of the species that causes visceral leishmaniasis (VL) in the Old World, while L. infantum chagasi (LIC) is present in the New World. Few studies address biological differences or the behavior of these strains during infection. These parasites live inside cells of their hosts, continuously evading microbicidal mechanisms and modulating the immune responses of these cells. One of the mechanisms used by these protozoa involves the L-arginine metabolism. Understanding the differences between Leishmania species and establishing an improved murine model for study of leishmaniasis are matters of extreme importance. Thereby, the objectives of this work were to analyze the biological and molecular differences between two Leishmania infantum strains (LII and LIC) and the degree of susceptibility to infection of mice with different genetic backgrounds. The infectivity in vivo and in vitro of LII and LIC strains was evaluated in BALB/c and Swiss Webster mice, as well the NOS and ARG activities. The LII strain was more infective than the LIC strain both in vivo and in vitro. In animals infected by the LII and LIC strains, differences in NOS and ARG activities occurred. In vitro, promastigotes of LII isolated from BALB/c and Swiss Webster mice showed higher ARG activity than LIC promastigotes during the growth curve. However, no difference was observed in intracellular NO production by promastigotes of these strains. The ARG gene sequences were compared, and those of both strains were identical. However, despite the similarity, the strains showed different expression levels of this gene. It can be concluded that although L. chagasi strains are considered identical to L. infantum strains from a molecular point of view, these strains have different biological behavior.
Collapse
Affiliation(s)
- Taiana Ferreira-Paes
- Laboratório de Bioquímica de Tripanosomatídeos, Instituto Oswaldo Cruz/Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Karen dos Santos Charret
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz/Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | - Raquel Ferreira Rodrigues
- Laboratório de Bioquímica de Tripanosomatídeos, Instituto Oswaldo Cruz/Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Leonor Laura Leon
- Laboratório de Bioquímica de Tripanosomatídeos, Instituto Oswaldo Cruz/Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
15
|
Cecílio P, Oristian J, Meneses C, Serafim TD, Valenzuela JG, Cordeiro da Silva A, Oliveira F. Engineering a vector-based pan-Leishmania vaccine for humans: proof of principle. Sci Rep 2020; 10:18653. [PMID: 33122717 PMCID: PMC7596519 DOI: 10.1038/s41598-020-75410-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022] Open
Abstract
Leishmaniasis is a spectrum of diseases transmitted by sand fly vectors that deposit Leishmania spp. parasites in the host skin during blood feeding. Currently, available treatment options are limited, associated with high toxicity and emerging resistance. Even though a vaccine for human leishmaniasis is considered an achievable goal, to date we still do not have one available, a consequence (amongst other factors) of a lack of pre-clinical to clinical translatability. Pre-exposure to uninfected sand fly bites or immunization with defined sand fly salivary proteins was shown to negatively impact infection. Still, cross-protection reports are rare and dependent on the phylogenetic proximity of the sand fly species, meaning that the applicability of a sand fly saliva-based vaccine will be limited to a defined geography, one parasite species and one form of leishmaniasis. As a proof of principle of a future vector saliva-based pan-Leishmania vaccine, we engineered through a reverse vaccinology approach that maximizes translation to humans, a fusion protein consisting of immunogenic portions of PdSP15 and LJL143, sand fly salivary proteins demonstrated as potential vaccine candidates against cutaneous and visceral leishmaniasis, respectively. The in silico analysis was validated ex vivo, through T cell proliferation experiments, proving that the fusion protein (administered as a DNA vaccine) maintained the immunogenicity of both PdSP15 and LJL143. Additionally, while no significant effect was detected in the context of L. major transmission by P. duboscqi, this DNA vaccine was defined as partially protective, in the context of L. major transmission by L. longipalpis sand flies. Importantly, a high IFNγ response alone was not enough to confer protection, that mainly correlated with low T cell mediated Leishmania-specific IL-4 and IL-10 responses, and consequently with high pro/anti-inflammatory cytokine ratios. Overall our immunogenicity data suggests that to design a potentially safe vector-based pan-Leishmania vaccine, without geographic restrictions and against all forms of leishmaniasis is an achievable goal. This is why we propose our approach as a proof-of principle, perhaps not only applicable to the anti-Leishmania vector-based vaccines' field, but also to other branches of knowledge that require the design of multi-epitope T cell vaccines with a higher potential for translation.
Collapse
Affiliation(s)
- Pedro Cecílio
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Parasite Disease Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto (FFUP), Porto, Portugal
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD, 20852, USA
| | - James Oristian
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD, 20852, USA
| | - Claudio Meneses
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD, 20852, USA
| | - Tiago D Serafim
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD, 20852, USA
| | - Jesus G Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD, 20852, USA
| | - Anabela Cordeiro da Silva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
- Parasite Disease Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.
- Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto (FFUP), Porto, Portugal.
| | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD, 20852, USA.
| |
Collapse
|
16
|
Santos MF, Alexandre-Pires G, Pereira MA, Gomes L, Rodrigues AV, Basso A, Reisinho A, Meireles J, Santos-Gomes GM, Pereira da Fonseca I. Immunophenotyping of Peripheral Blood, Lymph Node, and Bone Marrow T Lymphocytes During Canine Leishmaniosis and the Impact of Antileishmanial Chemotherapy. Front Vet Sci 2020; 7:375. [PMID: 32760744 PMCID: PMC7373748 DOI: 10.3389/fvets.2020.00375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/28/2020] [Indexed: 11/30/2022] Open
Abstract
Dogs are a major reservoir of Leishmania infantum, etiological agent of canine leishmaniosis (CanL) a zoonotic visceral disease of worldwide concern. Therapeutic protocols based on antileishmanial drugs are commonly used to treat sick dogs and improve their clinical condition. To better understand the impact of Leishmania infection and antileishmanial drugs on the dog's immune response, this study investigates the profile of CD4+ and CD8+ T cell subsets in peripheral blood, lymph node, and bone marrow of sick dogs and after two different CanL treatments. Two CanL groups of six dogs each were treated with either miltefosine or meglumine antimoniate combined with allopurinol. Another group of 10 clinically healthy dogs was used as control. Upon diagnosis and during the following 3 months of treatment, peripheral blood, popliteal lymph node, and bone marrow mononuclear cells were collected, labeled for surface markers CD45, CD3, CD4, CD8, CD25, and intracellular nuclear factor FoxP3, and T lymphocyte subpopulations were immunophenotyped by flow cytometry. CanL dogs presented an overall increased frequency of CD8+ and CD4+CD8+ double-positive T cells in all tissues and a decreased frequency of CD4+ T cells in the blood. Furthermore, there was a higher frequency of CD8+ T cells expressing CD25+FoxP3+ in the blood and bone marrow. During treatment, these subsets recovered to levels similar to those of healthy dogs. Nevertheless, antileishmanial therapy caused an increase of CD4+CD25+FoxP3+ T cells in all tissues, associated with the decrease of CD8+CD25−FoxP3− T cell percentages. These findings may support previous studies that indicate that L. infantum manipulates the dog's immune system to avoid the development of a protective response, ensuring the parasite's survival and the conditions that allow the completion of Leishmania life cycle. Both treatments used appear to have an effect on the dog's immune response, proving to be effective in promoting the normalization of T cell subsets.
Collapse
Affiliation(s)
- Marcos Ferreira Santos
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisbon, Portugal
| | - Graça Alexandre-Pires
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisbon, Portugal
| | - Maria A Pereira
- GHTM-Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova De Lisbon (UNL), Lisbon, Portugal
| | - Lídia Gomes
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisbon, Portugal
| | - Armanda V Rodrigues
- GHTM-Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova De Lisbon (UNL), Lisbon, Portugal
| | - Alexandra Basso
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Reisinho
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisbon, Portugal
| | - José Meireles
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisbon, Portugal
| | - Gabriela M Santos-Gomes
- GHTM-Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova De Lisbon (UNL), Lisbon, Portugal
| | - Isabel Pereira da Fonseca
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
17
|
Schrom EC, Levin SA, Graham AL. Quorum sensing via dynamic cytokine signaling comprehensively explains divergent patterns of effector choice among helper T cells. PLoS Comput Biol 2020; 16:e1008051. [PMID: 32730250 PMCID: PMC7392205 DOI: 10.1371/journal.pcbi.1008051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 06/13/2020] [Indexed: 12/13/2022] Open
Abstract
In the animal kingdom, various forms of swarming enable groups of autonomous individuals to transform uncertain information into unified decisions which are probabilistically beneficial. Crossing scales from individual to group decisions requires dynamically accumulating signals among individuals. In striking parallel, the mammalian immune system is also a group of decentralized autonomous units (i.e. cells) which collectively navigate uncertainty with the help of dynamically accumulating signals (i.e. cytokines). Therefore, we apply techniques of understanding swarm behavior to a decision-making problem in the mammalian immune system, namely effector choice among CD4+ T helper (Th) cells. We find that incorporating dynamic cytokine signaling into a simple model of Th differentiation comprehensively explains divergent observations of this process. The plasticity and heterogeneity of individual Th cells, the tunable mixtures of effector types that can be generated in vitro, and the polarized yet updateable group effector commitment often observed in vivo are all explained by the same set of underlying molecular rules. These rules reveal that Th cells harness dynamic cytokine signaling to implement a system of quorum sensing. Quorum sensing, in turn, may confer adaptive advantages on the mammalian immune system, especially during coinfection and during coevolution with manipulative parasites. This highlights a new way of understanding the mammalian immune system as a cellular swarm, and it underscores the power of collectives throughout nature.
Collapse
Affiliation(s)
- Edward C. Schrom
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, United States of America
- * E-mail:
| | - Simon A. Levin
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Andrea L. Graham
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, United States of America
| |
Collapse
|
18
|
Rawat AK, Pal K, Singh R, Anand A, Gupta S, Kishore D, Singh S, Singh RK. The CD200-CD200R cross-talk helps Leishmania donovani to down regulate macrophage and CD4 +CD44 + T cells effector functions in an NFκB independent manner. Int J Biol Macromol 2020; 151:394-401. [PMID: 32084478 DOI: 10.1016/j.ijbiomac.2020.02.189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/16/2020] [Accepted: 02/17/2020] [Indexed: 12/15/2022]
Abstract
The lacuna in the knowledge of immunobiology, especially in visceral infections that are fatal if left untreated, are a major hurdle in getting a vaccine candidate for leishmaniasis. Till date, only a few drugs are available to combat human leishmaniasis and a vaccine candidate either prophylactic or preventive is still awaited. Therefore, identification of host and parasitic factors involved in the regulation of specific immune mechanisms are essentially needed. In this study, we observed that CD200-CD200R immune inhibitory axis regulates host macrophages effectors properties and helps antigen experienced T cells (CD4+CD44+ T cells) to acquire anti-inflammatory cytokines (IL-4, IL-10, TGF-β, IL-27) producing abilities in an NFkB independent manner. After CD200 blocking the macrophages effectively inhibited proliferation of Leishmania amastigotes and also induced the production of IL-12, IFN-γ, TNF-α and nitric oxide (NOx). Further, the blocking of CD200 signaling also restored macrophages MHC-II expression and helped CD4+CD44+ T cells to produce pro-inflammatory cytokines like IL-2, IL-12 and IFN-γ. The finding of this study suggested the importance of immune inhibitory mechanisms in controlling Leishmania growth and survival and therefore, requires more studies to understand its role in vaccine induced immunity.
Collapse
Affiliation(s)
- Arun Kumar Rawat
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221 005, India
| | - Kavita Pal
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221 005, India
| | - Rajan Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221 005, India
| | - Anshul Anand
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221 005, India
| | - Smita Gupta
- Department of Microbiology, Institute of Medical Science, Banaras Hindu University, Varanasi 221 005, India
| | - Dhiraj Kishore
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221 005, India
| | - Sangram Singh
- Department of Biochemistry, Faculty of Science, Dr. RML Avadh University, Faizabad 224001, India
| | - Rakesh K Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221 005, India.
| |
Collapse
|
19
|
Mendes Costa D, Cecílio P, Santarém N, Cordeiro-da-Silva A, Tavares J. Murine infection with bioluminescent Leishmania infantum axenic amastigotes applied to drug discovery. Sci Rep 2019; 9:18989. [PMID: 31831809 PMCID: PMC6908656 DOI: 10.1038/s41598-019-55474-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/25/2019] [Indexed: 11/13/2022] Open
Abstract
Leishmaniasis is an important vector-borne neglected tropical disease caused by Leishmania parasites. Current anti-Leishmania chemotherapy is unsatisfactory, justifying the continued search for alternative treatment options. Herein, we demonstrate that luciferase-expressing Leishmania infantum axenic amastigotes, unlike promastigotes, are highly infectious to BALB/c mice and thus generate a robust bioluminescent signal in target organs, such as the liver and the spleen, as early as two weeks after infection. Treatment with the reference drugs amphotericin B and miltefosine was effective at reducing parasite burdens. This model allows the assessment of treatment efficacy using whole-mouse bioluminescence imaging without the need to wait several weeks for spleen infections to be detectable by this non-invasive method. In conclusion, we propose the use of this model in an initial approach to evaluate the treatment efficacy of promising chemical entities without having to sacrifice large numbers of animals or to wait several days for a readout.
Collapse
|
20
|
Brito CCB, da Silva HVC, Brondani DJ, de Faria AR, Ximenes RM, da Silva IM, de Albuquerque JFC, Castilho MS. Synthesis and biological evaluation of thiazole derivatives as LbSOD inhibitors. J Enzyme Inhib Med Chem 2019; 34:333-342. [PMID: 30734600 PMCID: PMC6327998 DOI: 10.1080/14756366.2018.1550752] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 10/09/2018] [Accepted: 11/18/2018] [Indexed: 11/08/2022] Open
Abstract
Leishmaniasis is considered as one of the major neglected tropical diseases due to its magnitude and wide geographic distribution. Leishmania braziliensis, responsible for cutaneous leishmaniasis, is the most prevalent species in Brazil. Superoxide dismutase (SOD) belongs to the antioxidant pathway of the parasites and human host. Despite the differences between SOD of Leishmania braziliensis and human make this enzyme a promising target for drug development efforts. No medicinal chemistry effort has been made to identify LbSOD inhibitors. Herein, we show that thermal shift assays (TSA) and fluorescent protein-labeled assays (FPLA) can be employed as primary and secondary screens to achieve this goal. Moreover, we show that thiazole derivatives bind to LbSOD with micromolar affinity.
Collapse
Affiliation(s)
- Camila C. Bitencourt Brito
- Programa de pós-graduação em Biotecnologia, Universidade Estadual de Feira de Santana, Feira de Santana, BA, Brazil
| | | | | | | | - Rafael Matos Ximenes
- Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | | | | | - Marcelo Santos Castilho
- Programa de pós-graduação em Biotecnologia, Universidade Estadual de Feira de Santana, Feira de Santana, BA, Brazil
- Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, BA, Brazil
| |
Collapse
|
21
|
Affiliation(s)
- Steven A. Frank
- Department of Ecology and Evolutionary Biology, University of California, Irvine, California, United States of America
| | | |
Collapse
|
22
|
Pérez-Cabezas B, Cecílio P, Gaspar TB, Gärtner F, Vasconcellos R, Cordeiro-da-Silva A. Understanding Resistance vs. Susceptibility in Visceral Leishmaniasis Using Mouse Models of Leishmania infantum Infection. Front Cell Infect Microbiol 2019; 9:30. [PMID: 30881923 PMCID: PMC6407322 DOI: 10.3389/fcimb.2019.00030] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 01/31/2019] [Indexed: 01/28/2023] Open
Abstract
Every year, up to 90,000 new cases of Visceral Leishmaniasis and 30,000 resultant deaths are estimated to occur worldwide. Such numbers give relevance to the continuous study of this complex form of the disease: a zoonosis and an anthroponosis; two known etiological agents (Leishmania infantum and L. donovani, respectively); with an estimated average ratio of 1 symptomatic per 10 asymptomatic individuals; and sometimes associated with atypical clinical presentations. This complexity, which results from a long co-evolutionary process involving vector-host, host-pathogen, and pathogen-vector interactions, is still not completely understood. The determinants of visceralization are not fully defined and the dichotomy resistance vs. susceptibility remains unsolved, translating into obstacles that delay the progress of global disease control. Inbred mouse models, with different susceptibility patterns to Leishmania infection, have been very useful in exploring this dichotomy. BALB/c and C57BL/6 mice were described as susceptible strains to L. donovani visceral infection, while SV/129 was considered resistant. Here, we used these three mouse models, but in the context of L. infantum infection, the other Leishmania species that cause visceral disease in humans, and dynamically compared their local and systemic infection-induced immune responses in order to establish a parallel and to ultimately better understand susceptibility vs. resistance in visceral leishmaniasis. Overall, our results suggest that C57BL/6 mice develop an intermediate “infection-phenotype” in comparison to BALB/c and SV/129 mouse strains, considering both the splenic parasite burden and the determined target organs weights. However, the immune mechanisms associated with the control of infection seem to be different in each mouse strain. We observed that both BALB/c and SV/129, but not C57BL/6 mice, show an infection-induced increase of splenic T follicular helper cells. On the other hand, differences detected in terms of CD21 expression by B cells early after infection, together with the quantified anti-Leishmania specific antibodies, suggest that SV/129 are faster than BALB/c and C57BL/6 mice in the assembly of an efficient B-cell response. Additionally, we observed an infection-induced increase in polyfunctional CD4+ T cells in the resistant SV/129 model, opposing an infection-induced increase in CD4+IL-10+ cells in susceptible BALB/c mice. Our data aligns with the observations reported for L. donovani infection and suggest that not only a single mechanism, but an interaction of several could be necessary for the control of this parasitic disease.
Collapse
Affiliation(s)
- Begoña Pérez-Cabezas
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Parasite Disease Group, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Pedro Cecílio
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Parasite Disease Group, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Tiago Bordeira Gaspar
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Cancer Signalling and Metabolism Group, Institute of Molecular Pathology and Immunology of University of Porto, Porto, Portugal.,Faculdade de Medicina, Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Fátima Gärtner
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Department of Molecular Pathology and Immunology, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal.,Glycobiology in Cancer Group, Institute of Molecular Pathology and Immunology of University of Porto, Universidade do Porto, Porto, Portugal
| | - Rita Vasconcellos
- Immunobiology Department, Biology Institute, Universidade Federal Fluminense, Rio de Janeiro, Brazil
| | - Anabela Cordeiro-da-Silva
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Parasite Disease Group, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| |
Collapse
|
23
|
Stamper BD, Davis M, Scott-Collins S, Tran J, Ton C, Simidyan A, Roberts SC. Model-based Evaluation of Gene Expression Changes in Response to Leishmania Infection. GENE REGULATION AND SYSTEMS BIOLOGY 2019; 13:1177625019828350. [PMID: 30792575 PMCID: PMC6376507 DOI: 10.1177/1177625019828350] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 12/21/2018] [Indexed: 12/16/2022]
Abstract
Since the development of high-density microarray technology in the late 1990s, global host gene expression changes in response to various stimuli have been extensively studied. More than a dozen peer-reviewed publications have investigated the effect of Leishmania infection in various models since 2001. This review covers the transcriptional changes in macrophage models induced by various Leishmania species and summarizes the resulting impact these studies have on our understanding of the host response to leishmaniasis in vitro. Characterization of the similarities and differences between various model systems will not only further our understanding of Leishmania-induced changes to macrophage gene expression but also identify potential therapeutic targets in the future.
Collapse
Affiliation(s)
| | - Madison Davis
- School of Pharmacy, Pacific University Oregon, Hillsboro, OR, USA
| | | | - Julie Tran
- School of Pharmacy, Pacific University Oregon, Hillsboro, OR, USA
| | - Caryn Ton
- School of Pharmacy, Pacific University Oregon, Hillsboro, OR, USA
| | - Agapi Simidyan
- School of Pharmacy, Pacific University Oregon, Hillsboro, OR, USA
| | - Sigrid C Roberts
- School of Pharmacy, Pacific University Oregon, Hillsboro, OR, USA
| |
Collapse
|
24
|
Goldberg MF, Roeske EK, Ward LN, Pengo T, Dileepan T, Kotov DI, Jenkins MK. Salmonella Persist in Activated Macrophages in T Cell-Sparse Granulomas but Are Contained by Surrounding CXCR3 Ligand-Positioned Th1 Cells. Immunity 2018; 49:1090-1102.e7. [PMID: 30552021 DOI: 10.1016/j.immuni.2018.10.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 08/16/2018] [Accepted: 10/10/2018] [Indexed: 12/11/2022]
Abstract
Salmonella enterica (Se) bacteria cause persistent intracellular infections while stimulating a robust interferon-γ-producing CD4+ T (Th1) cell response. We addressed this paradox of concomitant infection and immunity by tracking fluorescent Se organisms in mice. Se bacteria persisted in nitric oxide synthase (iNOS)-producing resident and recruited macrophages while inducing genes related to protection from nitric oxide. Se-infected cells occupied iNOS+ splenic granulomas that excluded T cells but were surrounded by mononuclear phagocytes producing the chemokines CXCL9 and CXCL10, and Se epitope-specific Th1 cells expressing CXCR3, the receptor for these chemokines. Blockade of CXCR3 inhibited Th1 occupancy of CXCL9/10-dense regions, reduced activation of the Th1 cells, and led to increased Se growth. Thus, intracellular Se bacteria survive in their hosts by counteracting toxic products of the innate immune response and by residing in T cell-sparse granulomas, away from abundant Th1 cells positioned via CXCR3 in a bordering region that act to limit infection.
Collapse
Affiliation(s)
- Michael F Goldberg
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Elizabeth K Roeske
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Lauren N Ward
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Thomas Pengo
- University of Minnesota Informatics Institute, Office of the Vice President for Research, Minneapolis, MN 55455, USA
| | - Thamotharampillai Dileepan
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Dmitri I Kotov
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Marc K Jenkins
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
25
|
Pérez-Cabezas B, Santarém N, Cecílio P, Silva C, Silvestre R, A M Catita J, Cordeiro da Silva A. More than just exosomes: distinct Leishmania infantum extracellular products potentiate the establishment of infection. J Extracell Vesicles 2018; 8:1541708. [PMID: 30455859 PMCID: PMC6237156 DOI: 10.1080/20013078.2018.1541708] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 09/21/2018] [Accepted: 10/23/2018] [Indexed: 12/19/2022] Open
Abstract
The use of secretion pathways for effector molecule delivery by microorganisms is a trademark of pathogenesis. Leishmania extracellular vesicles (EVs) were shown to have significant immunomodulatory potential. Still, they will act in conjunction with other released parasite-derived products that might modify the EVs effects. Notwithstanding, the immunomodulatory properties of these non-vesicular components and their influence in the infectious process remains unknown. To address this, we explored both in vitro and in vivo the immunomodulatory potential of promastigotes extracellular material (EXO), obtained as a whole or separated in two different fractions: EVs or vesicle depleted EXO (VDE). Using an air pouch model, we observed that EVs and VDE induced a dose-dependent cell recruitment profile different from the one obtained with parasites, attracting significantly fewer neutrophils and more dendritic cells (DCs). Additionally, when we co-inoculated parasites with extracellular products a drop in cell recruitment was observed. Moreover, in vitro, while VDE (but not EVs) downregulated the expression of DCs and macrophages activation markers, both products were able to diminish the responsiveness of these cells to LPS. Finally, the presence of Leishmania infantum extracellular products in the inoculum promoted a dose-dependent infection potentiation in vivo, highlighting their relevance for the infectious process. In conclusion, our data demonstrate that EVs are not the only relevant players among the parasite exogenous products. This, together with the dose-dependency observed, opens new avenues to the comprehension of Leishmania infectious process. The approach presented here should be exploited to revisit existing data and considered for future studies in other infection models.
Collapse
Affiliation(s)
- Begoña Pérez-Cabezas
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Parasite Disease Group, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Nuno Santarém
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Parasite Disease Group, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Pedro Cecílio
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Parasite Disease Group, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Cátia Silva
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Parasite Disease Group, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Ricardo Silvestre
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
| | - José A M Catita
- FP-ENAS Research Unit, UFP Energy, Environment and Health Research Unit, CEBIMED, Biomedical Research Centre, Fernando Pessoa University, Porto, Portugal.,Paralab, SA, Valbom, Portugal
| | - Anabela Cordeiro da Silva
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Parasite Disease Group, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto, Porto, Portugal
| |
Collapse
|
26
|
Bahrami F, Harandi AM, Rafati S. Biomarkers of Cutaneous Leishmaniasis. Front Cell Infect Microbiol 2018; 8:222. [PMID: 29998089 PMCID: PMC6029629 DOI: 10.3389/fcimb.2018.00222] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/11/2018] [Indexed: 12/12/2022] Open
Abstract
Cutaneous leishmaniasis (CL) is an immune-mediated skin pathology caused mainly by Leishmania (L.) major, Leishmania tropica, Leishmania braziliensis, L. mexicana, and L. amazonensis. The burden of CL in terms of morbidity and social stigmas are concentrated on certain developing countries in Asia, Africa, and South America. People with asymptomatic CL represent a large proportion of the infected individuals in the endemic areas who exhibit no lesion and can control the infection by as yet not fully understood mechanisms. Currently, there is no approved prophylactic control measure for CL. Discovery of biomarkers of CL infection and immunity can inform the development of more precise diagnostics tools as well as curative or preventive strategies to control CL. Herein, we provide a brief overview of the state-of-the-art for the biomarkers of CL with a special emphasis on the asymptomatic CL biomarkers. Among the identified CL biomarkers so far, direct biomarkers which indicate the actual presence of the infection as well as indirect biomarkers which reflect the host's reaction to the infection, such as alterations in delayed type hypersensitivity, T-cell subpopulations and cytokines, adenosine deaminase, and antibodies against the sand fly saliva proteins are discussed in detail. The future avenues such as the use of systems analysis to identify and characterize novel CL biomarkers are also discussed.
Collapse
Affiliation(s)
- Fariborz Bahrami
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Ali M Harandi
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sima Rafati
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
27
|
Diotallevi A, De Santi M, Buffi G, Ceccarelli M, Vitale F, Galluzzi L, Magnani M. Leishmania Infection Induces MicroRNA hsa-miR-346 in Human Cell Line-Derived Macrophages. Front Microbiol 2018; 9:1019. [PMID: 29867904 PMCID: PMC5966562 DOI: 10.3389/fmicb.2018.01019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/30/2018] [Indexed: 01/09/2023] Open
Abstract
Leishmaniasis is an anthropo-zoonotic disease caused by various Leishmania species. The clinical manifestations of the disease vary according to the species and host characteristics. Leishmania infection leads to subversion/modulation of the host’s innate immune response and cellular metabolic pathways. In the last years, it has been shown that many host cell gene expression and signaling pathways are targeted by Leishmania to subvert host defenses (e.g., oxidative damage, immune activation, antigen presentation, apoptosis) and allow parasite survival and replication. However, the molecular mechanisms triggered by the parasite are not fully elucidated. The role of miRNA has recently been evaluated in human or murine macrophages infected with Leishmania (Leishmania) major, L. (L.) donovani or L. (L.) amazonensis. However, no literature exists regarding miRNA dysregulation in host cells infected with L. (L.) infantum or L. (Viannia) species. Since we previously showed that L. (L.) infantum infection induced unfolded protein response (UPR) in macrophages, we focused on miR-346, which has been shown to be induced by the UPR-activated transcription factor sXBP1 and has a potential role in the modulation of the immune response. Macrophages differentiated from U937 and/or THP-1 human monocytic cells were infected with four L. (L.) infantum strain/clinical isolates and one L. (V.) sp. clinical isolate. A significant upregulation of miR-346 (p < 0.05) was observed in infections with all the Leishmania species tested. Moreover, RFX1 (a miR-346 predicted target gene) was found to be significantly downregulated (p < 0.05) after 48h infection, and miR-346 was found to have a role in this downregulation. The induction of miR-346 in macrophages infected with L. (L.) infantum and L. (V.) sp., reported here for the first time, could play a role in regulating macrophage functions since several MHC- or interferon-associated genes are among the targets of this miRNA. Hence, miR-346 could be considered an attractive anti-Leishmania drug target.
Collapse
Affiliation(s)
- Aurora Diotallevi
- Department of Biomolecular Sciences, Section of Biotechnology, University of Urbino, Fano, Italy
| | - Mauro De Santi
- Department of Biomolecular Sciences, Section of Hygiene, University of Urbino, Urbino, Italy
| | - Gloria Buffi
- Department of Biomolecular Sciences, Section of Biotechnology, University of Urbino, Fano, Italy
| | - Marcello Ceccarelli
- Department of Biomolecular Sciences, Section of Biotechnology, University of Urbino, Fano, Italy
| | - Fabrizio Vitale
- Istituto Zooprofilattico Sperimentale della Sicilia, Palermo, Italy
| | - Luca Galluzzi
- Department of Biomolecular Sciences, Section of Biotechnology, University of Urbino, Fano, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, Section of Biotechnology, University of Urbino, Fano, Italy
| |
Collapse
|
28
|
Almeida APMM, Machado LFM, Doro D, Nascimento FC, Damasceno L, Gazzinelli RT, Fernandes AP, Junqueira C. New Vaccine Formulations Containing a Modified Version of the Amastigote 2 Antigen and the Non-Virulent Trypanosoma cruzi CL-14 Strain Are Highly Antigenic and Protective against Leishmania infantum Challenge. Front Immunol 2018; 9:465. [PMID: 29599776 PMCID: PMC5863692 DOI: 10.3389/fimmu.2018.00465] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 02/21/2018] [Indexed: 12/21/2022] Open
Abstract
Visceral leishmaniasis (VL) is a major public health issue reported as the second illness in mortality among all tropical diseases. Clinical trials have shown that protection against VL is associated with robust T cell responses, especially those producing IFN-γ. The Leishmania amastigote 2 (A2) protein has been repeatedly described as immunogenic and protective against VL in different animal models; it is recognized by human T cells, and it is also commercially available in a vaccine formulation containing saponin against canine VL. Moving toward a more appropriate formulation for human vaccination, here, we tested a new optimized version of the recombinant protein (rA2), designed for Escherichia coli expression, in combination with adjuvants that have been approved for human use. Moreover, aiming at improving the cellular immune response triggered by rA2, we generated a recombinant live vaccine vector using Trypanosoma cruzi CL-14 non-virulent strain, named CL-14 A2. Mice immunized with respective rA2, adsorbed in Alum/CpG B297, a TLR9 agonist recognized by mice and human homologs, or with the recombinant CL-14 A2 parasites through homologous prime-boost protocol, were evaluated for antigen-specific immune responses and protection against Leishmania infantum promastigote challenge. Immunization with the new rA2/Alum/CpG formulations and CL-14 A2 transgenic vectors elicited stronger cellular immune responses than control groups, as shown by increased levels of IFN-γ, conferring protection against L. infantum challenge. Interestingly, the use of the wild-type CL-14 alone was enough to boost immunity and confer protection, confirming the previously reported immunogenic potential of this strain. Together, these results support the success of both the newly designed rA2 antigen and the ability of T. cruzi CL-14 to induce strong T cell-mediated immune responses against VL in animal models when used as a live vaccine vector. In conclusion, the vaccination strategies explored here reveal promising alternatives for the development of new rA2 vaccine formulations to be translated human clinical trials.
Collapse
Affiliation(s)
- Ana Paula M M Almeida
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leopoldo F M Machado
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Daniel Doro
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Frederico C Nascimento
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Ricardo Tostes Gazzinelli
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil.,Division of Infectious Disease, University of Massachusetts Medical School, Worcester, MA, United States
| | - Ana Paula Fernandes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | |
Collapse
|
29
|
Soulat D, Bogdan C. Function of Macrophage and Parasite Phosphatases in Leishmaniasis. Front Immunol 2017; 8:1838. [PMID: 29312331 PMCID: PMC5743797 DOI: 10.3389/fimmu.2017.01838] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 12/05/2017] [Indexed: 01/23/2023] Open
Abstract
The kinetoplastid protozoan parasites belonging to the genus Leishmania are the causative agents of different clinical forms of leishmaniasis, a vector-borne infectious disease with worldwide prevalence. The protective host immune response against Leishmania parasites relies on myeloid cells such as dendritic cells and macrophages in which upon stimulation by cytokines (e.g., interferon-γ) a complex network of signaling pathways is switched on leading to strong antimicrobial activities directed against the intracellular parasite stage. The regulation of these pathways classically depends on post-translational modifications of proteins, with phosphorylation events playing a cardinal role. Leishmania parasites deactivate their phagocytic host cells by inducing specific mammalian phosphatases that are capable to impede signaling. On the other hand, there is now also evidence that Leishmania spp. themselves express phosphatases that might target host cell molecules and thereby facilitate the intracellular survival of the parasite. This review will present an overview on the modulation of host phosphatases by Leishmania parasites as well as on the known families of Leishmania phosphatases and their possible function as virulence factors. A more detailed understanding of the role of phosphatases in Leishmania–host cell interactions might open new avenues for the treatment of non-healing, progressive forms of leishmaniasis.
Collapse
Affiliation(s)
- Didier Soulat
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany.,Medical Immunology Campus Erlangen, Interdisciplinary Center of the FAU, Erlangen, Germany
| | - Christian Bogdan
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany.,Medical Immunology Campus Erlangen, Interdisciplinary Center of the FAU, Erlangen, Germany
| |
Collapse
|
30
|
Cecílio P, Pérez-Cabezas B, Fernández L, Moreno J, Carrillo E, Requena JM, Fichera E, Reed SG, Coler RN, Kamhawi S, Oliveira F, Valenzuela JG, Gradoni L, Glueck R, Gupta G, Cordeiro-da-Silva A. Pre-clinical antigenicity studies of an innovative multivalent vaccine for human visceral leishmaniasis. PLoS Negl Trop Dis 2017; 11:e0005951. [PMID: 29176865 PMCID: PMC5720812 DOI: 10.1371/journal.pntd.0005951] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 12/07/2017] [Accepted: 09/11/2017] [Indexed: 01/18/2023] Open
Abstract
The notion that previous infection by Leishmania spp. in endemic areas leads to robust anti-Leishmania immunity, supports vaccination as a potentially effective approach to prevent disease development. Nevertheless, to date there is no vaccine available for human leishmaniasis. We optimized and assessed in vivo the safety and immunogenicity of an innovative vaccine candidate against human visceral leishmaniasis (VL), consisting of Virus-Like Particles (VLP) loaded with three different recombinant proteins (LJL143 from Lutzomyia longipalpis saliva as the vector-derived (VD) component, and KMP11 and LeishF3+, as parasite-derived (PD) antigens) and adjuvanted with GLA-SE, a TLR4 agonist. No apparent adverse reactions were observed during the experimental time-frame, which together with the normal hematological parameters detected seems to point to the safety of the formulation. Furthermore, measurements of antigen-specific cellular and humoral responses, generally higher in immunized versus control groups, confirmed the immunogenicity of the vaccine formulation. Interestingly, the immune responses against the VD protein were reproducibly more robust than those elicited against leishmanial antigens, and were apparently not caused by immunodominance of the VD antigen. Remarkably, priming with the VD protein alone and boosting with the complete vaccine candidate contributed towards an increase of the immune responses to the PD antigens, assessed in the form of increased ex vivo CD4+ and CD8+ T cell proliferation against both the PD antigens and total Leishmania antigen (TLA). Overall, our immunogenicity data indicate that this innovative vaccine formulation represents a promising anti-Leishmania vaccine whose efficacy deserves to be tested in the context of the "natural infection".
Collapse
Affiliation(s)
- Pedro Cecílio
- Parasite Disease group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- IBMC—Instituto de Biologia Celular e Molecular, Universidade do Porto, Porto, Portugal
- Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto, Porto, Portugal
| | - Begoña Pérez-Cabezas
- Parasite Disease group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- IBMC—Instituto de Biologia Celular e Molecular, Universidade do Porto, Porto, Portugal
| | - Laura Fernández
- WHO Collaborating Centre for Leishmaniasis, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Moreno
- WHO Collaborating Centre for Leishmaniasis, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - Eugenia Carrillo
- WHO Collaborating Centre for Leishmaniasis, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - José M. Requena
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Epifanio Fichera
- Etna Biotech S.R.L, via Vincenzo Lancia, 57—Zona Industriale Blocco Palma 1, Catania, Italy
| | - Steven G. Reed
- Infectious Disease Research Institute (IDRI), Seattle, WA, United States of America
| | - Rhea N. Coler
- Infectious Disease Research Institute (IDRI), Seattle, WA, United States of America
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, NIAID, NIH, Rockville, MD, United States of America
| | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, NIAID, NIH, Rockville, MD, United States of America
| | - Jesus G. Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, NIAID, NIH, Rockville, MD, United States of America
| | - Luigi Gradoni
- Unit of Vector-borne Diseases and International Health, Istituto Superiore di Sanità, Rome, Italy
| | - Reinhard Glueck
- Etna Biotech S.R.L, via Vincenzo Lancia, 57—Zona Industriale Blocco Palma 1, Catania, Italy
| | - Gaurav Gupta
- Etna Biotech S.R.L, via Vincenzo Lancia, 57—Zona Industriale Blocco Palma 1, Catania, Italy
| | - Anabela Cordeiro-da-Silva
- Parasite Disease group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- IBMC—Instituto de Biologia Celular e Molecular, Universidade do Porto, Porto, Portugal
- Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto, Porto, Portugal
| |
Collapse
|
31
|
Gonçalves-de-Albuquerque SDC, Pessoa-e-Silva R, Trajano-Silva LAM, de Goes TC, de Morais RCS, da C. Oliveira CN, de Lorena VMB, de Paiva-Cavalcanti M. The Equivocal Role of Th17 Cells and Neutrophils on Immunopathogenesis of Leishmaniasis. Front Immunol 2017; 8:1437. [PMID: 29163510 PMCID: PMC5670345 DOI: 10.3389/fimmu.2017.01437] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 10/16/2017] [Indexed: 01/17/2023] Open
Abstract
Advances in the understanding of leishmaniasis progression indicate that cellular interactions more complex than the Th1/Th2 paradigm define the course of infection. Th17 cells are a crucial modulator of adaptive immunity against Leishmania parasites acting mainly on neutrophil recruitment and playing a dual role at the site of infection. This review describes the roles of both these cell types in linking innate defense responses to the establishment of specific immunity. We focus on the Th17-neutrophil interaction as a crucial component of anti-Leishmania immunity, and the clinical evolution of cutaneous or visceral leishmaniasis. To date, information obtained through experimental models and patient evaluations suggests that the influence of the presence of interleukin (IL)-17 (the main cytokine produced by Th17 cells) and neutrophils during Leishmania infections is strictly dependent on the tissue (skin or liver/spleen) and parasite species. Also, the time at which neutrophils are recruited, and the persistence of IL-17 in the infection microenvironment, may also be significant. A clearer understanding of these interactions will enable better measurement of the influence of IL-17 and its regulators, and contribute to the identification of disease/resistance biomarkers.
Collapse
Affiliation(s)
| | - Rômulo Pessoa-e-Silva
- Department of Microbiology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | - Lays A. M. Trajano-Silva
- Department of Microbiology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | - Tayná Correia de Goes
- Department of Microbiology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | - Rayana C. S. de Morais
- Department of Microbiology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | - Cíntia N. da C. Oliveira
- Department of Microbiology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | - Virgínia M. B. de Lorena
- Department of Microbiology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | - Milena de Paiva-Cavalcanti
- Department of Microbiology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| |
Collapse
|
32
|
Vivarini ÁDC, Calegari-Silva TC, Saliba AM, Boaventura VS, França-Costa J, Khouri R, Dierckx T, Dias-Teixeira KL, Fasel N, Barral AMP, Borges VM, Van Weyenbergh J, Lopes UG. Systems Approach Reveals Nuclear Factor Erythroid 2-Related Factor 2/Protein Kinase R Crosstalk in Human Cutaneous Leishmaniasis. Front Immunol 2017; 8:1127. [PMID: 28959260 PMCID: PMC5605755 DOI: 10.3389/fimmu.2017.01127] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 08/28/2017] [Indexed: 01/15/2023] Open
Abstract
Leishmania parasites infect macrophages, causing a wide spectrum of human diseases, from cutaneous to visceral forms. In search of novel therapeutic targets, we performed comprehensive in vitro and ex vivo mapping of the signaling pathways upstream and downstream of antioxidant transcription factor [nuclear factor erythroid 2-related factor 2 (Nrf2)] in cutaneous leishmaniasis (CL), by combining functional assays in human and murine macrophages with a systems biology analysis of in situ (skin biopsies) CL patient samples. First, we show the PKR pathway controls the expression and activation of Nrf2 in Leishmania amazonensis infection in vitro. Nrf2 activation also required PI3K/Akt signaling and autophagy mechanisms. Nrf2- or PKR/Akt-deficient macrophages exhibited increased levels of ROS/RNS and reduced expression of Sod1 Nrf2-dependent gene and reduced parasite load. L. amazonensis counteracted the Nrf2 inhibitor Keap1 through the upregulation of p62 via PKR. This Nrf2/Keap1 observation was confirmed in situ in skin biopsies from Leishmania-infected patients. Next, we explored the ex vivo transcriptome in CL patients, as compared to healthy controls. We found the antioxidant response element/Nrf2 signaling pathway was significantly upregulated in CL, including downstream target p62. In silico enrichment analysis confirmed upstream signaling by interferon and PI3K/Akt, and validated our in vitro findings. Our integrated in vitro, ex vivo, and in silico approach establish Nrf2 as a central player in human cutaneous leishmaniasis and reveal Nrf2/PKR crosstalk and PI3K/Akt pathways as potential therapeutic targets.
Collapse
Affiliation(s)
- Áislan de Carvalho Vivarini
- Laboratory of Molecular Parasitology, Carlos Chagas Filho Biophysics Institute, Center of Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Teresa Cristina Calegari-Silva
- Laboratory of Molecular Parasitology, Carlos Chagas Filho Biophysics Institute, Center of Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandra Mattos Saliba
- Department of Microbiology, Immunology and Parasitology - FCM/UERJ, State University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Viviane Sampaio Boaventura
- Integrated Laboratory of Microbiology and Immunoregulation, Gonçalo Moniz Research Center, Oswaldo Cruz Foundation, Salvador, Bahia, Brazil
| | - Jaqueline França-Costa
- Integrated Laboratory of Microbiology and Immunoregulation, Gonçalo Moniz Research Center, Oswaldo Cruz Foundation, Salvador, Bahia, Brazil
| | - Ricardo Khouri
- Integrated Laboratory of Microbiology and Immunoregulation, Gonçalo Moniz Research Center, Oswaldo Cruz Foundation, Salvador, Bahia, Brazil
| | - Tim Dierckx
- Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Karina Luiza Dias-Teixeira
- Laboratory of Molecular Parasitology, Carlos Chagas Filho Biophysics Institute, Center of Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nicolas Fasel
- Faculty of Biology and Medicine, Department of Biochemistry, University of Lausanne, Lausanne, Switzerland
| | - Aldina Maria Prado Barral
- Integrated Laboratory of Microbiology and Immunoregulation, Gonçalo Moniz Research Center, Oswaldo Cruz Foundation, Salvador, Bahia, Brazil
| | - Valéria Matos Borges
- Integrated Laboratory of Microbiology and Immunoregulation, Gonçalo Moniz Research Center, Oswaldo Cruz Foundation, Salvador, Bahia, Brazil
| | - Johan Van Weyenbergh
- Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Ulisses Gazos Lopes
- Laboratory of Molecular Parasitology, Carlos Chagas Filho Biophysics Institute, Center of Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
33
|
Belo R, Santarém N, Pereira C, Pérez-Cabezas B, Macedo F, Leite-de-Moraes M, Cordeiro-da-Silva A. Leishmania infantum Exoproducts Inhibit Human Invariant NKT Cell Expansion and Activation. Front Immunol 2017; 8:710. [PMID: 28674535 PMCID: PMC5474685 DOI: 10.3389/fimmu.2017.00710] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 06/01/2017] [Indexed: 01/28/2023] Open
Abstract
Leishmania infantum is one of the major parasite species associated with visceral leishmaniasis, a severe form of the disease that can become lethal if untreated. This obligate intracellular parasite has developed diverse strategies to escape the host immune response, such as exoproducts (Exo) carrying a wide range of molecules, including parasite virulence factors, which are potentially implicated in early stages of infection. Herein, we report that L. infantum Exo and its two fractions composed of extracellular vesicles (EVs) and vesicle-depleted-exoproducts (VDEs) inhibit human peripheral blood invariant natural killer T (iNKT) cell expansion in response to their specific ligand, the glycolipid α-GalactosylCeramide (α-GalCer), as well as their capacity to promptly produce IL-4 and IFNγ. Using plate-bound CD1d and α-GalCer, we found that Exo, EV, and VDE fractions reduced iNKT cell activation in a dose-dependent manner, suggesting that they prevented α-GalCer presentation by CD1d molecules. This direct effect on CD1d was confirmed by the observation that CD1d:α-GalCer complex formation was impaired in the presence of Exo, EV, and VDE fractions. Furthermore, lipid extracts from the three compounds mimicked the inhibition of iNKT cell activation. These lipid components of L. infantum exoproducts, including EV and VDE fractions, might compete for CD1-binding sites, thus blocking iNKT cell activation. Overall, our results provide evidence for a novel strategy through which L. infantum can evade immune responses of mammalian host cells by preventing iNKT lymphocytes from recognizing glycolipids in a TCR-dependent manner.
Collapse
Affiliation(s)
- Renata Belo
- Parasite Disease Group, Institute for Molecular and Cell Biology (IBMC), Institute for Investigation and Innovation in Health (i3S), Porto, Portugal.,Laboratory of Immunoregulation and Immunopathology, Institut Necker-Enfants Malades, CNRS UMR 8253 and INSERM UMR 1151, Paris, France.,Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Nuno Santarém
- Parasite Disease Group, Institute for Molecular and Cell Biology (IBMC), Institute for Investigation and Innovation in Health (i3S), Porto, Portugal
| | - Cátia Pereira
- Cell Activation and Gene Expression, Institute for Molecular and Cell Biology (IBMC), Institute for Investigation and Innovation in Health (i3S), Porto, Portugal
| | - Begoña Pérez-Cabezas
- Parasite Disease Group, Institute for Molecular and Cell Biology (IBMC), Institute for Investigation and Innovation in Health (i3S), Porto, Portugal
| | - Fátima Macedo
- Cell Activation and Gene Expression, Institute for Molecular and Cell Biology (IBMC), Institute for Investigation and Innovation in Health (i3S), Porto, Portugal.,Department of Medical Science, Aveiro University, Aveiro, Portugal
| | - Maria Leite-de-Moraes
- Laboratory of Immunoregulation and Immunopathology, Institut Necker-Enfants Malades, CNRS UMR 8253 and INSERM UMR 1151, Paris, France.,Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Anabela Cordeiro-da-Silva
- Parasite Disease Group, Institute for Molecular and Cell Biology (IBMC), Institute for Investigation and Innovation in Health (i3S), Porto, Portugal.,Faculty of Pharmacy, Department of Biological Sciences, University of Porto, Porto, Portugal
| |
Collapse
|
34
|
Silva JC, Zacarias DA, Silva VC, Rolão N, Costa DL, Costa CH. Comparison of optical microscopy and quantitative polymerase chain reaction for estimating parasitaemia in patients with kala-azar and modelling infectiousness to the vector Lutzomyia longipalpis. Mem Inst Oswaldo Cruz 2017; 111:517-22. [PMID: 27439033 PMCID: PMC4981120 DOI: 10.1590/0074-02760160185] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 06/06/2016] [Indexed: 12/04/2022] Open
Abstract
Currently, the only method for identifying infective hosts with Leishmania infantum to the vector Lutzomyia longipalpis is xenodiagnosis. More recently, quantitative polymerase chain reaction (qPCR) has been used to model human reservoir competence by assuming that detection of parasite DNA indicates the presence of viable parasites for infecting vectors. Since this assumption has not been proven, this study aimed to verify this hypothesis. The concentration of amastigotes in the peripheral blood of 30 patients with kala-azar was microscopically verified by leukoconcentration and was compared to qPCR estimates. Parasites were identified in 4.8 mL of peripheral blood from 67% of the patients, at a very low concentration (average 0.3 parasites/mL). However, qPCR showed 93% sensitivity and the estimated parasitaemia was over a thousand times greater, both in blood and plasma, with higher levels in plasma than in blood. Furthermore, the microscopic count of circulating parasites and the qPCR parasitaemia estimates were not mathematically compatible with the published proportions of infected sandflies in xenodiagnostic studies. These findings suggest that qPCR does not measure the concentration of circulating parasites, but rather measures DNA from other sites, and that blood might not be the main source of infection for vectors.
Collapse
Affiliation(s)
- Jailthon C Silva
- Universidade Federal do Piauí, Departamento de Medicina Comunitária, Instituto de Doenças Tropicais Natan Portella, Laboratório de Leishmanioses, Teresina, PI, Brasil
| | - Danielle A Zacarias
- Universidade Federal do Piauí, Departamento de Medicina Comunitária, Instituto de Doenças Tropicais Natan Portella, Laboratório de Leishmanioses, Teresina, PI, Brasil
| | - Vladimir C Silva
- Universidade Federal do Piauí, Departamento de Medicina Comunitária, Instituto de Doenças Tropicais Natan Portella, Laboratório de Leishmanioses, Teresina, PI, Brasil
| | - Nuno Rolão
- Universidade Nova de Lisboa, Instituto de Higiene e Medicina Tropical, Lisboa, Portugal
| | - Dorcas L Costa
- Universidade Federal do Piauí, Departamento Materno-Infantil, Teresina, PI, Brasil
| | - Carlos Hn Costa
- Universidade Federal do Piauí, Departamento de Medicina Comunitária, Instituto de Doenças Tropicais Natan Portella, Laboratório de Leishmanioses, Teresina, PI, Brasil
| |
Collapse
|
35
|
Celes FS, Trovatti E, Khouri R, Van Weyenbergh J, Ribeiro SJL, Borges VM, Barud HS, de Oliveira CI. DETC-based bacterial cellulose bio-curatives for topical treatment of cutaneous leishmaniasis. Sci Rep 2016; 6:38330. [PMID: 27922065 PMCID: PMC5138610 DOI: 10.1038/srep38330] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 11/07/2016] [Indexed: 11/09/2022] Open
Abstract
The treatment of leishmaniasis still relies on drugs with potentially serious adverse effects. Herein, we tested a topical formulation of bacterial cellulose (BC) membranes containing Diethyldithiocarbamate (DETC), a superoxide dismutase 1 inhibitor. Leishmania-infected macrophages exposed to BC-DETC resulted in parasite killing, without pronounced toxic effects to host cells. This outcome was associated with lower SOD1 activity and higher production of superoxide and cytokine mediators. Topical application of BC-DETC significantly decreased lesion size, parasite load and the inflammatory response at the infection site, as well as the production of both IFN-γ and TNF. Combination of topical BC-DETC plus intraperitoneal Sbv also significantly reduced disease development and parasite load. The leishmanicidal effect of BC-DETC was extended to human macrophages infected with L. braziliensis, highlighting the feasibility of BC-DETC as a topical formulation for chemotherapy of cutaneous leishmaniasis caused by L. braziliensis.
Collapse
Affiliation(s)
| | - Eliane Trovatti
- Instituto de Química, Universidade Estadual Paulista, Araraquara, SP, Brazil.,Universidade de Araraquara-UNIARA, Araraquara, SP, Brazil
| | | | - Johan Van Weyenbergh
- Rega Institute for Medical Research, Department of Microbiology and Immunology, K. U. Leuven, Belgium
| | - Sidney J L Ribeiro
- Instituto de Química, Universidade Estadual Paulista, Araraquara, SP, Brazil
| | | | - Hernane S Barud
- Instituto de Química, Universidade Estadual Paulista, Araraquara, SP, Brazil.,Universidade de Araraquara-UNIARA, Araraquara, SP, Brazil
| | - Camila I de Oliveira
- Instituto Gonçalo Moniz, FIOCRUZ, Salvador, BA, Brazil.,Instituto de Investigação em Imunologia (iii), INCT, São Paulo, Brazil
| |
Collapse
|
36
|
Pérez-Cabezas B, Cecílio P, Robalo AL, Silvestre R, Carrillo E, Moreno J, San Martín JV, Vasconcellos R, Cordeiro-da-Silva A. Interleukin-27 Early Impacts Leishmania infantum Infection in Mice and Correlates with Active Visceral Disease in Humans. Front Immunol 2016; 7:478. [PMID: 27867384 PMCID: PMC5095612 DOI: 10.3389/fimmu.2016.00478] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 10/20/2016] [Indexed: 12/26/2022] Open
Abstract
The complexity of Leishmania–host interactions, one of the main leishmaniasis issues, is yet to be fully understood. We detected elevated IL-27 plasma levels in European patients with active visceral disease caused by Leishmania infantum, which returned to basal levels after successful treatment, suggesting this cytokine as a probable infection mediator. We further addressed this hypothesis recurring to two classical susceptible visceral leishmaniasis mouse models. BALB/c, but not C57BL/6 mice, showed increased IL-27 systemic levels after infection, which was associated with an upregulation of IL-27p28 expression by dendritic cells and higher parasite burdens. Neutralization of IL-27 in acutely infected BALB/c led to decreased parasite burdens and a transient increase in IFN-γ+ splenic T cells, while administration of IL-27 to C57BL/6 promoted a local anti-inflammatory cytokine response at the site of infection and increased parasite loads. Overall, we show that, as in humans, BALB/c IL-27 systemic levels are infection dependently upregulated and may favor parasite installation by controlling inflammation.
Collapse
Affiliation(s)
- Begoña Pérez-Cabezas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Pedro Cecílio
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Ana Luisa Robalo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Ricardo Silvestre
- ICVS - Instituto de Investigação em Ciências da Vida e Saúde, Escola de Ciências da Saúde, Universidade do Minho, Braga, Portugal; ICVS/3B's - Laboratório Associado, Braga, Portugal
| | - Eugenia Carrillo
- WHO Collaborating Centre for Leishmaniasis, Centro Nacional de Microbiología, Instituto de Salud Carlos III , Madrid , Spain
| | - Javier Moreno
- WHO Collaborating Centre for Leishmaniasis, Centro Nacional de Microbiología, Instituto de Salud Carlos III , Madrid , Spain
| | | | - Rita Vasconcellos
- Departamento de Imunobiologia, Instituto de Biologia, Universidade Federal Fluminense , Niterói , Brazil
| | - Anabela Cordeiro-da-Silva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal; Faculdade de Farmácia, Departamento de Ciências Biológicas, Universidade do Porto, Porto, Portugal
| |
Collapse
|
37
|
Gupta P, Srivastav S, Saha S, Das PK, Ukil A. Leishmania donovani inhibits macrophage apoptosis and pro-inflammatory response through AKT-mediated regulation of β-catenin and FOXO-1. Cell Death Differ 2016; 23:1815-1826. [PMID: 27662364 DOI: 10.1038/cdd.2016.101] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 07/18/2016] [Accepted: 08/17/2016] [Indexed: 12/25/2022] Open
Abstract
In order to establish infection, intra-macrophage parasite Leishmania donovani needs to inhibit host defense parameters like inflammatory cytokine production and apoptosis. In the present study, we demonstrate that the parasite achieves both by exploiting a single host regulator AKT for modulating its downstream transcription factors, β-catenin and FOXO-1. L. donovani-infected RAW264.7 and bone marrow-derived macrophages (BMDM) treated with AKT inhibitor or dominant negative AKT constructs showed decreased anti-inflammatory cytokine production and increased host cell apoptosis resulting in reduced parasite survival. Infection-induced activated AKT triggered phosphorylation-mediated deactivation of its downstream target, GSK-3β. Inactivated GSK-3β, in turn, could no longer sequester cytosolic β-catenin, an anti-apoptotic transcriptional regulator, as evidenced from its nuclear translocation during infection. Constitutively active GSK-3β-transfected L. donovani-infected cells mimicked the effects of AKT inhibition and siRNA-mediated silencing of β-catenin led to disruption of mitochondrial potential along with increased caspase-3 activity and IL-12 production leading to decreased parasite survival. In addition to activating anti-apoptotic β-catenin, phospho-AKT inhibits activation of FOXO-1, a pro-apoptotic transcriptional regulator. Nuclear retention of FOXO-1, inhibited during infection, was reversed when infected cells were transfected with dominant negative AKT constructs. Overexpression of FOXO-1 in infected macrophages not only documented increased apoptosis but promoted enhanced TLR4 expression and NF-κB activity along with an increase in IL-1β and decrease in IL-10 secretion. In vivo administration of AKT inhibitor significantly decreased liver and spleen parasite burden and switched cytokine balance in favor of host. In contrast, GSK-3β inhibitor did not result in any significant change in infectivity parameters. Collectively our findings revealed that L. donovani triggered AKT activation to regulate GSK-3β/β-catenin/FOXO-1 axis, thus ensuring inhibition of both host cell apoptosis and immune response essential for its intra-macrophage survival.
Collapse
Affiliation(s)
- Purnima Gupta
- Department of Biochemistry, University of Calcutta, Kolkata 700019, India
| | - Supriya Srivastav
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Shriya Saha
- Department of Biochemistry, University of Calcutta, Kolkata 700019, India
| | - Pijush K Das
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Anindita Ukil
- Department of Biochemistry, University of Calcutta, Kolkata 700019, India
| |
Collapse
|
38
|
Figueiredo ABD, Souza-Testasicca MC, Afonso LCC. Purinergic signaling and infection by Leishmania: A new approach to evasion of the immune response. Biomed J 2016; 39:244-250. [PMID: 27793266 PMCID: PMC6139394 DOI: 10.1016/j.bj.2016.08.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/18/2016] [Accepted: 08/02/2016] [Indexed: 12/20/2022] Open
Abstract
Infection by protozoan parasites is part of the most common Tropical Neglected Diseases. In the case of leishmaniasis, several millions of people are at risk of contracting the disease. In spite of innumerous studies that elucidated the immune response capable of killing the parasite, the understanding of the evasion mechanisms utilized by the parasite to survive within the very cell responsible for its destruction is still incomplete. In this review, we offer a new approach to the control of the immune response against the parasite. The ability of the parasite to modulate the levels of extracellular ATP and adenosine either by directly acting on the levels of these molecules or by inducing the expression of CD39 and CD73 on the infected cell may influence the magnitude of the immune response against the parasite contributing to its growth and survival.
Collapse
Affiliation(s)
- Amanda Braga de Figueiredo
- Immunoparasitology Laboratory, Department of Biological Sciences, ICEB/NUPEB, Federal University of Ouro Preto, Brazil
| | | | - Luis Carlos Crocco Afonso
- Immunoparasitology Laboratory, Department of Biological Sciences, ICEB/NUPEB, Federal University of Ouro Preto, Brazil.
| |
Collapse
|
39
|
Roy S, Mandal C. Leishmania donovani Utilize Sialic Acids for Binding and Phagocytosis in the Macrophages through Selective Utilization of Siglecs and Impair the Innate Immune Arm. PLoS Negl Trop Dis 2016; 10:e0004904. [PMID: 27494323 PMCID: PMC4975436 DOI: 10.1371/journal.pntd.0004904] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 07/14/2016] [Indexed: 12/15/2022] Open
Abstract
Background Leishmania donovani, belonging to a unicellular protozoan parasite, display the differential level of linkage-specific sialic acids on their surface. Sialic acids binding immunoglobulin-like lectins (siglecs) are a class of membrane-bound receptors present in the haematopoetic cell lineages interact with the linkage-specific sialic acids. Here we aimed to explore the utilization of sialic acids by Leishmania donovani for siglec-mediated binding, phagocytosis, modulation of innate immune response and signaling pathways for establishment of successful infection in the host. Methodology/Principle Findings We have found enhanced binding of high sialic acids containing virulent strains (AG83+Sias) with siglec-1 and siglec-5 present on macrophages compared to sialidase treated AG83+Sias (AG83-Sias) and low sialic acids-containing avirulent strain (UR6) by flow cytometry. This specific receptor-ligand interaction between sialic acids and siglecs were further confirmed by confocal microscopy. Sialic acids-siglec-1-mediated interaction of AG83+Sias with macrophages induced enhanced phagocytosis. Additionally, sialic acids-siglec-5 interaction demonstrated reduced ROS, NO generation and Th2 dominant cytokine response upon infection with AG83+Sias in contrast to AG83-Sias and UR6. Sialic acids-siglecs binding also facilitated multiplication of intracellular amastigotes. Moreover, AG83+Sias induced sialic acids-siglec-5-mediated upregulation of host phosphatase SHP-1. Such sialic acids-siglec interaction was responsible for further downregulation of MAPKs (p38, ERK and JNK) and PI3K/Akt pathways followed by the reduced translocation of p65 subunit of NF-κβ to the nucleus from cytosol in the downstream signaling pathways. This sequence of events was reversed in AG83-Sias and UR6-infected macrophages. Besides, siglec-knockdown macrophages also showed the reversal of AG83+Sias infection-induced effector functions and downstream signaling events. Conclusions/Significances Taken together, this study demonstrated that virulent parasite (AG83+Sias) establish a unique sialic acids-mediated binding and subsequent phagocytosis in the host cell through the selective exploitation of siglec-1. Additionally, sialic acids-siglec-5 interaction altered the downstream signaling pathways which contributed impairment of immune effector functions of macrophages. To the best of our knowledge, this is a comprehensive report describing sialic acids-siglec interactions and their role in facilitating uptake of the virulent parasite within the host. Sialic acids are nine carbon sugars present on terminal cell surface glycoproteins and glycolipids. Siglec is a membrane receptor that belongs to an immunoglobulin super family present in almost all the haematopoetic cell lineages. There are 14 different types of siglecs present on human immune cells that take an active part in balancing the magnitude of immunological reactions. In general, these siglecs bind with sialic acids and negatively regulate the immune response. Leishmania contains sialic acids on its surface. Virulent parasites utilize this sugar to bind with macrophages through siglec-1 and siglec-5 compared to low sialic acids containing avirulent parasites. Such sialic acids-siglec-mediated interactions exhibited a suppressed host immune response which helped them to establish successful infection compared to desialylated virulent and avirulent parasites, as well as, siglec-depleted macrophages. Interestingly, interaction between sialic acids and siglec-1 induced enhanced phagocytosis, while sialic acids-siglec-5 interaction upregulated the phosphatase SHP-1. This interaction with the virulent strain exhibited deactivation of various downstream signaling pathways and ultimately controlled translocation of a functional component of transcription factor NF-κβ for regulation of cytokines and other effector molecules in infected macrophages. Thus, the interaction between the parasite and the host cells through sialic acids-siglec binding is clearly a newly identified mechanism by which parasites can establish successful infection by subverting the host’s innate immune response.
Collapse
Affiliation(s)
- Saptarshi Roy
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Chitra Mandal
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- * E-mail:
| |
Collapse
|
40
|
Anand S, Madhubala R. Twin Attributes of Tyrosyl-tRNA Synthetase of Leishmania donovani: A HOUSEKEEPING PROTEIN TRANSLATION ENZYME AND A MIMIC OF HOST CHEMOKINE. J Biol Chem 2016; 291:17754-71. [PMID: 27382051 DOI: 10.1074/jbc.m116.727107] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Indexed: 12/13/2022] Open
Abstract
Aminoacyl-tRNA synthetases (aaRSs) are housekeeping enzymes essential for protein synthesis. Apart from their parent aminoacylation activity, several aaRSs perform non-canonical functions in diverse biological processes. The present study explores the twin attributes of Leishmania tyrosyl-tRNA synthetase (LdTyrRS) namely, aminoacylation, and as a mimic of host CXC chemokine. Leishmania donovani is a protozoan parasite. Its genome encodes a single copy of tyrosyl-tRNA synthetase. We first tested the canonical aminoacylation role of LdTyrRS. The recombinant protein was expressed, and its kinetic parameters were determined by aminoacylation assay. To study the physiological role of LdTyrRS in Leishmania, gene deletion mutations were attempted via targeted gene replacement. The heterozygous mutants showed slower growth kinetics and exhibited attenuated virulence. LdTyrRS appears to be an essential gene as the chromosomal null mutants did not survive. Our data also highlights the non-canonical function of L. donovani tyrosyl-tRNA synthetase. We show that LdTyrRS protein is present in the cytoplasm and exits from the parasite cytoplasm into the extracellular medium. The released LdTyrRS functions as a neutrophil chemoattractant. We further show that LdTyrRS specifically binds to host macrophages with its ELR (Glu-Leu-Arg) peptide motif. The ELR-CXCR2 receptor interaction mediates this binding. This interaction triggers enhanced secretion of the proinflammatory cytokines TNF-α and IL-6 by host macrophages. Our data indicates a possible immunomodulating role of LdTyrRS in Leishmania infection. This study provides a platform to explore LdTyrRS as a potential target for drug development.
Collapse
Affiliation(s)
- Sneha Anand
- From the School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rentala Madhubala
- From the School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| |
Collapse
|
41
|
Geiger A, Bossard G, Sereno D, Pissarra J, Lemesre JL, Vincendeau P, Holzmuller P. Escaping Deleterious Immune Response in Their Hosts: Lessons from Trypanosomatids. Front Immunol 2016; 7:212. [PMID: 27303406 PMCID: PMC4885876 DOI: 10.3389/fimmu.2016.00212] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/17/2016] [Indexed: 12/21/2022] Open
Abstract
The Trypanosomatidae family includes the genera Trypanosoma and Leishmania, protozoan parasites displaying complex digenetic life cycles requiring a vertebrate host and an insect vector. Trypanosoma brucei gambiense, Trypanosoma cruzi, and Leishmania spp. are important human pathogens causing human African trypanosomiasis (HAT or sleeping sickness), Chagas' disease, and various clinical forms of Leishmaniasis, respectively. They are transmitted to humans by tsetse flies, triatomine bugs, or sandflies, and affect millions of people worldwide. In humans, extracellular African trypanosomes (T. brucei) evade the hosts' immune defenses, allowing their transmission to the next host, via the tsetse vector. By contrast, T. cruzi and Leishmania sp. have developed a complex intracellular lifestyle, also preventing several mechanisms to circumvent the host's immune response. This review seeks to set out the immune evasion strategies developed by the different trypanosomatids resulting from parasite-host interactions and will focus on: clinical and epidemiological importance of diseases; life cycles: parasites-hosts-vectors; innate immunity: key steps for trypanosomatids in invading hosts; deregulation of antigen-presenting cells; disruption of efficient specific immunity; and the immune responses used for parasite proliferation.
Collapse
Affiliation(s)
- Anne Geiger
- UMR INTERTRYP, IRD-CIRAD, CIRAD TA A-17/G, Montpellier, France
| | | | - Denis Sereno
- UMR INTERTRYP, IRD-CIRAD, CIRAD TA A-17/G, Montpellier, France
| | - Joana Pissarra
- UMR INTERTRYP, IRD-CIRAD, CIRAD TA A-17/G, Montpellier, France
| | | | - Philippe Vincendeau
- UMR 177, IRD-CIRAD Université de Bordeaux Laboratoire de Parasitologie, Bordeaux, France
| | - Philippe Holzmuller
- UMRCMAEE CIRAD-INRA TA-A15/G “Contrôle des maladies animales exotiques et émergentes”, Montpellier, France
| |
Collapse
|
42
|
Mukbel RM, Khasharmeh RH, Hijjawi NS, Khalifeh MS, Hatmal MM, McDowell MA. Human immune response to salivary proteins of wild-caught Phlebotomus papatasi. Parasitol Res 2016; 115:3345-55. [PMID: 27160331 DOI: 10.1007/s00436-016-5094-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 04/26/2016] [Indexed: 12/15/2022]
Abstract
Phlebotomine sand flies are the known vectors of Leishmania parasites. New approaches in vaccination against Leishmania have investigated the possibility of integrating Phlebotomus papatasi salivary proteins to enhance the immune response and protect against the transmission of the infection. The aim of the present study was to screen human immune responses to wild sand fly saliva and evaluate immunogenic salivary proteins. Blood samples were collected from donors in control and sand fly infested areas. Antibodies specific for sand fly antigens in donor plasma were probed using immunoblotting. In addition, recall proliferation capability of peripheral blood mononuclear cells (PBMC) was tested after sand fly salivary homogenates stimulation. The significant immunogenic salivary proteins (SPs) identified by immunoblotting were SP28, SP32, and SP36. A specific proliferative response of PBMC after stimulation with sand fly salivary homogenates was evident in donors that have antibody responses against sand fly salivary proteins. Individuals with antibody recognition to a higher number of salivary proteins (i.e., 3 or more SP bands) showed lower PBMC proliferative responses after in vitro stimulation with salivary gland homogenates (SGH) only in the sand fly infested, leishmaniasis free area. Interestingly, the presence of a humoral immune response to many SP antigens inversely correlates with a strong cell-mediated immune response (CMI). It was also noticed that some other heavily expressed antigens, in sand fly salivary homogenate, lack or have weak humoral immune reactivity in exposed individuals. Therefore, considering these antigens alone as CMI activators, without including the immunodominant humoral immune response proteins, needs future investigation.
Collapse
Affiliation(s)
- Rami M Mukbel
- Department of Basic Veterinary Medical Science, Faculty of Veterinary Medicine, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan.
| | - Rehab H Khasharmeh
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, The Hashemite University, PO Box 150459, Zarqa, 13115, Jordan
| | - Nawal S Hijjawi
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, The Hashemite University, PO Box 150459, Zarqa, 13115, Jordan
| | - Mohammed S Khalifeh
- Department of Basic Veterinary Medical Science, Faculty of Veterinary Medicine, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| | - Ma'mon M Hatmal
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, The Hashemite University, PO Box 150459, Zarqa, 13115, Jordan
| | - Mary Ann McDowell
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA
| |
Collapse
|
43
|
HIV-1 Tat protein enhances the intracellular growth of Leishmania amazonensis via the ds-RNA induced protein PKR. Sci Rep 2015; 5:16777. [PMID: 26608746 PMCID: PMC4660360 DOI: 10.1038/srep16777] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 10/15/2015] [Indexed: 02/06/2023] Open
Abstract
HIV-1 co-infection with human parasitic diseases is a growing public health problem worldwide. Leishmania parasites infect and replicate inside macrophages, thereby subverting host signaling pathways, including the response mediated by PKR. The HIV-1 Tat protein interacts with PKR and plays a pivotal role in HIV-1 replication. This study shows that Tat increases both the expression and activation of PKR in Leishmania-infected macrophages. Importantly, the positive effect of Tat addition on parasite growth was dependent on PKR signaling, as demonstrated in PKR-deficient macrophages or macrophages treated with the PKR inhibitor. The effect of HIV-1 Tat on parasite growth was prevented when the supernatant of HIV-1-infected macrophages was treated with neutralizing anti-HIV-1 Tat prior to Leishmania infection. The addition of HIV-1 Tat to Leishmania-infected macrophages led to inhibition of iNOS expression, modulation of NF-kB activation and enhancement of IL-10 expression. Accordingly, the expression of a Tat construct containing mutations in the basic region (49–57aa), which is responsible for the interaction with PKR, favored neither parasite growth nor IL-10 expression in infected macrophages. In summary, we show that Tat enhances Leishmania growth through PKR signaling.
Collapse
|
44
|
Singha UK, Hamilton V, Chaudhuri M. Tim62, a Novel Mitochondrial Protein in Trypanosoma brucei, Is Essential for Assembly and Stability of the TbTim17 Protein Complex. J Biol Chem 2015; 290:23226-39. [PMID: 26240144 DOI: 10.1074/jbc.m115.663492] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Indexed: 11/06/2022] Open
Abstract
Trypanosoma brucei, the causative agent of human African trypanosomiasis, possesses non-canonical mitochondrial protein import machinery. Previously, we characterized the essential translocase of the mitochondrial inner membrane (TIM) consisting of Tim17 in T. brucei. TbTim17 is associated with TbTim62. Here we show that TbTim62, a novel protein, is localized in the mitochondrial inner membrane, and its import into mitochondria depends on TbTim17. Knockdown (KD) of TbTim62 decreased the steady-state levels of TbTim17 post-transcriptionally. Further analysis showed that import of TbTim17 into mitochondria was not inhibited, but its half-life was reduced >4-fold due to TbTim62 KD. Blue-native gel electrophoresis revealed that TbTim62 is present primarily in ∼150-kDa and also in ∼1100-kDa protein complexes, whereas TbTim17 is present in multiple complexes within the range of ∼300 to ∼1100 kDa. TbTim62 KD reduced the levels of both TbTim62 as well as TbTim17 protein complexes. Interestingly, TbTim17 was accumulated as lower molecular mass complexes in TbTim62 KD mitochondria. Furthermore, depletion of TbTim62 hampered the assembly of the ectopically expressed TbTim17-2X-myc into TbTim17 protein complex. Co-immunoprecipitation analysis revealed that association of TbTim17 with mHSP70 was markedly reduced in TbTim62 KD mitochondria. All together our results demonstrate that TbTim62, a unique mitochondrial protein in T. brucei, is required for the formation of a stable TbTim17 protein complex. TbTim62 KD destabilizes this complex, and unassembled TbTim17 degrades. Therefore, TbTim62 acts as a novel regulatory factor to maintain the levels of TIM in T. brucei mitochondria.
Collapse
Affiliation(s)
- Ujjal K Singha
- From the Department of Microbiology and Immunology, Meharry Medical College, Nashville, Tennessee 37208
| | - VaNae Hamilton
- From the Department of Microbiology and Immunology, Meharry Medical College, Nashville, Tennessee 37208
| | - Minu Chaudhuri
- From the Department of Microbiology and Immunology, Meharry Medical College, Nashville, Tennessee 37208
| |
Collapse
|
45
|
Ali N, Nakhasi HL, Valenzuela JG, Reis AB. Targeted Immunology for Prevention and Cure of VL. Front Immunol 2014; 5:660. [PMID: 25566268 PMCID: PMC4271696 DOI: 10.3389/fimmu.2014.00660] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 12/08/2014] [Indexed: 12/21/2022] Open
Affiliation(s)
- Nahid Ali
- Infectious Diseases and Immunology, Indian Institute of Chemical Biology , Kolkata , India
| | - Hira L Nakhasi
- US Food and Drug Administration , Silver Spring, MD , USA
| | - Jesus G Valenzuela
- National Institute of Allergy and Infectious Diseases , Rockville, MD , USA
| | | |
Collapse
|