1
|
Carpinter BA, Renhe DC, Bellei JCB, Vieira CD, Rodolphi CM, Ferreira MVR, de Freitas CS, Neto AFDS, Coelho EAF, Mietto BDS, Gomes FLR, Rocha VN, Scopel KKG. DHA-rich fish oil plays a protective role against experimental cerebral malaria by controlling inflammatory and mechanical events from infection. J Nutr Biochem 2024; 123:109492. [PMID: 37866427 DOI: 10.1016/j.jnutbio.2023.109492] [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: 02/04/2023] [Revised: 05/17/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
Every year, thousands of children, particularly those under 5 years old, die because of cerebral malaria (CM). Following conventional treatment, approximately 25% of surviving individuals have lifelong severe neurocognitive sequelae. Therefore, improved conventional therapies or effective alternative therapies that prevent the severe infection are crucial. Omega-3 (Ω-3) polyunsaturated fatty acids (PUFAs) are known to have antioxidative and anti-inflammatory effects and protect against diverse neurological disorders, including Alzheimer's and Parkinson's diseases. However, little is known regarding the effects of Ω-3 PUFAs against parasitic infections. In this study, C57BL/6 mice received supplemental treatment of a fish oil rich in the Ω-3 PUFA, docosahexaenoic acid (DHA), which was started 15 days prior to infection with Plasmodium berghei ANKA and was maintained until the end of the study. Animals treated with the highest doses of DHA, 3.0 and 6.0 g/kg body weight, had 60 and 80% chance of survival, respectively, while all nontreated mice died by the 7th day postinfection due to CM. Furthermore, the parasite load during the critical period for CM development (5th to 11th day postinfection) was controlled in treated mice. However, after this period all animals developed high levels of parasitemia until the 20th day of infection. DHA treatment also effectively reduced blood-brain barrier (BBB) damage and brain edema and completely prevented brain hemorrhage and vascular occlusion. A strong anti-inflammatory profile was observed in the brains of DHA-treated mice, as well as, an increased number of neutrophil and reduced number of CD8+ T leukocytes in the spleen. Thus, this is the first study to demonstrate that the prophylactic use of DHA-rich fish oil exerts protective effects against experimental CM, reducing the mechanical and immunological events caused by the P. berghei ANKA infection.
Collapse
Affiliation(s)
- Bárbara Albuquerque Carpinter
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Daniela Chaves Renhe
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Jéssica Correa Bezerra Bellei
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Carolina David Vieira
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Cinthia Magalhães Rodolphi
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | | | - Camila Simões de Freitas
- Post-graduation Program in Health Sciences, Infectology and Tropical Medicine, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Adolfo Firmino da Silva Neto
- Department of Biology, Research Centre of Cellular Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Eduardo Antônio Ferraz Coelho
- Post-graduation Program in Health Sciences, Infectology and Tropical Medicine, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Bruno de Siqueira Mietto
- Department of Biology, Research Centre of Cellular Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | | | - Vinicius Novaes Rocha
- Department of Veterinary Medicine, Research Centre of Pathology and Veterinary Histology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Kézia Katiani Gorza Scopel
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil.
| |
Collapse
|
2
|
Leroux M, Luquain-Costaz C, Lawton P, Azzouz-Maache S, Delton I. Fatty Acid Composition and Metabolism in Leishmania Parasite Species: Potential Biomarkers or Drug Targets for Leishmaniasis? Int J Mol Sci 2023; 24:ijms24054702. [PMID: 36902138 PMCID: PMC10003364 DOI: 10.3390/ijms24054702] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/22/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Fatty acids have received growing interest in Leishmania biology with the characterization of the enzymes allowing the complete fatty acid synthesis of this trypanosomatid parasite. This review presents a comparative analysis of the fatty acid profiles of the major classes of lipids and phospholipids in different species of Leishmania with cutaneous or visceral tropism. Specificities relating to the parasite forms, resistance to antileishmanial drugs, and host/parasite interactions are described as well as comparisons with other trypanosomatids. Emphasis is placed on polyunsaturated fatty acids and their metabolic and functional specificities, in particular, their conversion into oxygenated metabolites that are inflammatory mediators able to modulate metacyclogenesis and parasite infectivity. The impact of lipid status on the development of leishmaniasis and the potential of fatty acids as therapeutic targets or candidates for nutritional interventions are discussed.
Collapse
Affiliation(s)
- Marine Leroux
- CNRS 5007, LAGEPP, Université of Lyon, Université Claude Bernard Lyon 1, 69100 Villeurbanne, France
| | - Céline Luquain-Costaz
- CNRS 5007, LAGEPP, Université of Lyon, Université Claude Bernard Lyon 1, 69100 Villeurbanne, France
- Department of Biosciences, INSA Lyon, 69100 Villeurbanne, France
| | - Philippe Lawton
- CNRS 5007, LAGEPP, Université of Lyon, Université Claude Bernard Lyon 1, 69100 Villeurbanne, France
| | - Samira Azzouz-Maache
- CNRS 5007, LAGEPP, Université of Lyon, Université Claude Bernard Lyon 1, 69100 Villeurbanne, France
| | - Isabelle Delton
- CNRS 5007, LAGEPP, Université of Lyon, Université Claude Bernard Lyon 1, 69100 Villeurbanne, France
- Department of Biosciences, INSA Lyon, 69100 Villeurbanne, France
- Correspondence:
| |
Collapse
|
3
|
Rebouças-Silva J, Amorim NA, Jesus-Santos FH, de Lima JA, Lima JB, Berretta AA, Borges VM. Leishmanicidal and immunomodulatory properties of Brazilian green propolis extract (EPP-AF ®) and a gel formulation in a pre-clinical model. Front Pharmacol 2023; 14:1013376. [PMID: 36843932 PMCID: PMC9949379 DOI: 10.3389/fphar.2023.1013376] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Leishmaniasis is a widespread group of neglected vector-borne tropical diseases that possess serious therapeutic limitations. Propolis has been extensively used in traditional medical applications due to its range of biological effects, including activity against infectious agents. Here we evaluated the leishmanicidal and immunomodulatory properties of Brazilian green propolis extract (EPP-AF®) and a gel formulation incorporating EPP-AF®, in both in vitro and in vivo models of Leishmania amazonensis infection. Propolis extract, obtained from a standardized blend following hydroalcoholic extraction, showed the characteristic fingerprint of Brazilian green propolis as confirmed by HPLC/DAD. A carbopol 940 gel formulation was obtained containing propolis glycolic extract at 3.6% w/w. The release profile, assessed using the Franz diffusion cell protocol, demonstrated a gradual and prolonged release of p-coumaric acid and artepillin C from the carbomer gel matrix. Quantification of p-coumaric acid and artepillin C in the gel formulation over time revealed that p-coumaric acid followed the Higuchi model, dependent on the disintegration of the pharmaceutical preparation, while artepillin C followed a zero-order profile with sustained release. In vitro analysis revealed the ability of EPP-AF® to reduce the infection index of infected macrophages (p < 0.05), while also modulating the production of inflammatory biomarkers. Decreases in nitric oxide and prostaglandin E2 levels were observed (p < 0.01), suggesting low iNOS and COX-2 activity. Furthermore, EPP-AF® treatment was found to induce heme oxygenase-1 antioxidant enzyme expression in both uninfected and L. amazonensis-infected cells, as well as inhibit IL-1β production in infected cells (p < 0.01). ERK-1/2 phosphorylation was positively correlated with TNF-α production (p < 0.05), yet no impact on parasite load was detected. In vivo analysis indicated the effectiveness of topical treatment with EPP-AF® gel alone (p < 0.05 and p < 0.01), or in combination with pentavalent antimony (p < 0.05 and p < 0.001), in the reduction of lesion size in the ears of L. amazonensis-infected BALB/c mice after seven or 3 weeks of treatment, respectively. Taken together, the present results reinforce the leishmanicidal and immunomodulatory effects of Brazilian green propolis, and demonstrate promising potential for the EPP-AF® propolis gel formulation as a candidate for adjuvant therapy in the treatment of Cutaneous Leishmaniasis.
Collapse
Affiliation(s)
- Jéssica Rebouças-Silva
- Laboratory of Inflammation and Biomarkers, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia, Brazil,Faculty of Medicine of Bahia, Federal University of Bahia (UFBA), Salvador, Bahia, Brazil
| | - Nathaly Alcazar Amorim
- Laboratory of Research, Development and Innovation, Apis Flora Industrial e Comercial Ltda, Ribeirão Preto, São Paulo, Brazil
| | - Flávio Henrique Jesus-Santos
- Laboratory of Inflammation and Biomarkers, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia, Brazil,Faculty of Medicine of Bahia, Federal University of Bahia (UFBA), Salvador, Bahia, Brazil
| | - Jéssica Aparecida de Lima
- Laboratory of Research, Development and Innovation, Apis Flora Industrial e Comercial Ltda, Ribeirão Preto, São Paulo, Brazil
| | | | - Andresa A. Berretta
- Laboratory of Research, Development and Innovation, Apis Flora Industrial e Comercial Ltda, Ribeirão Preto, São Paulo, Brazil,*Correspondence: Andresa A. Berretta, ; Valéria M. Borges,
| | - Valéria M. Borges
- Laboratory of Inflammation and Biomarkers, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia, Brazil,Faculty of Medicine of Bahia, Federal University of Bahia (UFBA), Salvador, Bahia, Brazil,*Correspondence: Andresa A. Berretta, ; Valéria M. Borges,
| |
Collapse
|
4
|
Silva RCMC, Vasconcelos LR, Travassos LH. The different facets of heme-oxygenase 1 in innate and adaptive immunity. Cell Biochem Biophys 2022; 80:609-631. [PMID: 36018440 DOI: 10.1007/s12013-022-01087-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 07/20/2022] [Indexed: 11/26/2022]
Abstract
Heme oxygenase (HO) enzymes are responsible for the main oxidative step in heme degradation, generating equimolar amounts of free iron, biliverdin and carbon monoxide. HO-1 is induced as a crucial stress response protein, playing protective roles in physiologic and pathological conditions, due to its antioxidant, anti-apoptotic and anti-inflammatory effects. The mechanisms behind HO-1-mediated protection are being explored by different studies, affecting cell fate through multiple ways, such as reduction in intracellular levels of heme and ROS, transcriptional regulation, and through its byproducts generation. In this review we focus on the interplay between HO-1 and immune-related signaling pathways, which culminate in the activation of transcription factors important in immune responses and inflammation. We also discuss the dual interaction of HO-1 and inflammatory mediators that govern resolution and tissue damage. We highlight the dichotomy of HO-1 in innate and adaptive immune cells development and activation in different disease contexts. Finally, we address different known anti-inflammatory pharmaceuticals that are now being described to modulate HO-1, and the possible contribution of HO-1 in their anti-inflammatory effects.
Collapse
Affiliation(s)
- Rafael Cardoso Maciel Costa Silva
- Laboratory of Immunoreceptors and Signaling, Instituto de Biofísica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Luiz Ricardo Vasconcelos
- Cellular Signaling and Cytoskeletal Function Laboratory, The Francis Crick Institute, London, UK
| | - Leonardo Holanda Travassos
- Laboratory of Immunoreceptors and Signaling, Instituto de Biofísica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
5
|
Nascimento MT, Cordeiro RSO, Abreu C, Santos CP, Peixoto F, Duarte GA, Cardoso T, de Oliveira CI, Carvalho E, Carvalho LP. Pioglitazone, a Peroxisome Proliferator-Activated Receptor-γ Agonist, Downregulates the Inflammatory Response in Cutaneous Leishmaniasis Patients Without Interfering in Leishmania braziliensis Killing by Monocytes. Front Cell Infect Microbiol 2022; 12:884237. [PMID: 35909958 PMCID: PMC9329526 DOI: 10.3389/fcimb.2022.884237] [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: 02/25/2022] [Accepted: 06/13/2022] [Indexed: 11/16/2022] Open
Abstract
Patients with cutaneous leishmaniasis (CL) due to Leishmania braziliensis infection have an exacerbated inflammatory response associated with tissue damage and ulcer development. An increase in the rate of patients who fail therapy with pentavalent antimony has been documented. An adjuvant therapy with an anti-inflammatory drug with the potential of Leishmania killing would benefit CL patients. The aim of the present study was to investigate the contribution of peroxisome proliferator-activated receptor-γ (PPAR-γ) activation by pioglitazone in the regulation of the inflammatory response and L. braziliensis killing by monocytes. Pioglitazone is an oral drug used in the treatment of diabetes, and its main mechanism of action is through the activation of PPAR-γ, which is expressed in many cell types of the immune response. We found that activation of PPAR-γ by pioglitazone decreases the inflammatory response in CL patients without affecting L. braziliensis killing by monocytes. Our data suggest that pioglitazone may serve as an adjunctive treatment for CL caused by L. braziliensis.
Collapse
Affiliation(s)
- Maurício T. Nascimento
- Laboratório de Pesquisas Clínicas, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
- Serviço de Imunologia, Complexo Hospitalar Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Ravena S. O. Cordeiro
- Laboratório de Pesquisas Clínicas, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
| | - Cayo Abreu
- Laboratório de Pesquisas Clínicas, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
| | - Camila P. Santos
- Laboratório de Pesquisas Clínicas, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Fábio Peixoto
- Laboratório de Pesquisas Clínicas, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Gabriela A. Duarte
- Laboratório de Enfermidades Infecciosas Transmitidas por Vetores, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
| | - Thiago Cardoso
- Laboratório de Pesquisas Clínicas, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
- Serviço de Imunologia, Complexo Hospitalar Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Camila I. de Oliveira
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
- Laboratório de Enfermidades Infecciosas Transmitidas por Vetores, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
- Ministério de Ciências e Tecnologia, Instituto Nacional de Ciências e Tecnologia-Doenças Tropicais, Salvador, Brazil
| | - Edgar M. Carvalho
- Laboratório de Pesquisas Clínicas, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
- Serviço de Imunologia, Complexo Hospitalar Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
- Ministério de Ciências e Tecnologia, Instituto Nacional de Ciências e Tecnologia-Doenças Tropicais, Salvador, Brazil
| | - Lucas P. Carvalho
- Laboratório de Pesquisas Clínicas, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
- Serviço de Imunologia, Complexo Hospitalar Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
- Ministério de Ciências e Tecnologia, Instituto Nacional de Ciências e Tecnologia-Doenças Tropicais, Salvador, Brazil
| |
Collapse
|
6
|
Malta-Santos H, Fukutani KF, Sorgi CA, Queiroz ATL, Nardini V, Silva J, Lago A, Carvalho LP, Machado PLR, Bozza PT, França-Costa J, Faccioli LH, Carvalho EM, Andrade BB, Borges VM. Multi-omic Analyses of Plasma Cytokines, Lipidomics, and Transcriptomics Distinguish Treatment Outcomes in Cutaneous Leishmaniasis. iScience 2020; 23:101840. [PMID: 33313489 PMCID: PMC7721649 DOI: 10.1016/j.isci.2020.101840] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/09/2020] [Accepted: 11/18/2020] [Indexed: 12/13/2022] Open
Abstract
Leishmania braziliensis infection frequently results in cutaneous leishmaniasis (CL). An increase in incidence of drug-resistant CL leading to treatment failure has been reported. Identification of reliable predictors of treatment outcomes is necessary to optimize patient care. Here, we performed a prospective case-control study in which plasma levels of cytokines and lipid mediators were assessed at different time points during antileishmanial therapy in patients with CL from Brazil. Multidimensional analyses were employed to describe a combination of biomarkers able to predict and characterize treatment failure. We found a biosignature influenced mainly by plasma levels of lipid mediators that accurately predicted treatment failure. Furthermore, transcriptomic analysis of a publicly available data set revealed that expression levels of genes related to lipid metabolism measured in skin lesions could distinguish treatment outcomes in CL. Thus, activation of pathways linked to lipid biosynthesis predicts treatment failure in CL. The biomarkers identified may be further explored as therapeutic targets.
Collapse
Affiliation(s)
- Hayna Malta-Santos
- Faculdade de Medicina da Bahia (FAMED), Universidade Federal da Bahia, Salvador, Brazil.,Instituto Gonçalo Moniz (IGM), Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
| | - Kiyoshi F Fukutani
- Instituto Gonçalo Moniz (IGM), Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER), Salvador, Brazil
| | - Carlos A Sorgi
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto (FCFRP-USP), Universidade de São Paulo (USP), São Paulo, Brazil
| | - Artur T L Queiroz
- Instituto Gonçalo Moniz (IGM), Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER), Salvador, Brazil
| | - Viviane Nardini
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto (FCFRP-USP), Universidade de São Paulo (USP), São Paulo, Brazil
| | - Juliana Silva
- Serviço de Imunologia, C-HUPES, Universidade Federal da Bahia, Salvador, Brazil
| | - Alex Lago
- Serviço de Imunologia, C-HUPES, Universidade Federal da Bahia, Salvador, Brazil
| | - Lucas P Carvalho
- Instituto Gonçalo Moniz (IGM), Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Serviço de Imunologia, C-HUPES, Universidade Federal da Bahia, Salvador, Brazil
| | - Paulo L R Machado
- Serviço de Imunologia, C-HUPES, Universidade Federal da Bahia, Salvador, Brazil
| | - Patrícia T Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Jaqueline França-Costa
- Instituto Gonçalo Moniz (IGM), Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Serviço de Imunologia, C-HUPES, Universidade Federal da Bahia, Salvador, Brazil
| | - Lucia H Faccioli
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto (FCFRP-USP), Universidade de São Paulo (USP), São Paulo, Brazil
| | - Edgar M Carvalho
- Faculdade de Medicina da Bahia (FAMED), Universidade Federal da Bahia, Salvador, Brazil.,Instituto Gonçalo Moniz (IGM), Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Serviço de Imunologia, C-HUPES, Universidade Federal da Bahia, Salvador, Brazil
| | - Bruno B Andrade
- Faculdade de Medicina da Bahia (FAMED), Universidade Federal da Bahia, Salvador, Brazil.,Instituto Gonçalo Moniz (IGM), Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER), Salvador, Brazil.,Escola Bahiana de Medicina e Saúde Pública, Salvador, Brazil.,Universidade Salvador (UNIFACS), Laureate Universities, Salvador, Brazil
| | - Valéria M Borges
- Faculdade de Medicina da Bahia (FAMED), Universidade Federal da Bahia, Salvador, Brazil.,Instituto Gonçalo Moniz (IGM), Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
| |
Collapse
|
7
|
Saunders EC, McConville MJ. Immunometabolism of Leishmania granulomas. Immunol Cell Biol 2020; 98:832-844. [PMID: 32780446 DOI: 10.1111/imcb.12394] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/06/2020] [Accepted: 08/06/2020] [Indexed: 12/19/2022]
Abstract
Leishmania are parasitic protists that cause a spectrum of diseases in humans characterized by the formation of granulomatous lesions in the skin or other tissues, such as liver and spleen. The extent to which Leishmania granulomas constrain or promote parasite growth is critically dependent on the host T-helper type 1/T-helper type 2 immune response and the localized functional polarization of infected and noninfected macrophages toward a classically (M1) or alternatively (M2) activated phenotype. Recent studies have shown that metabolic reprograming of M1 and M2 macrophages underpins the capacity of these cells to act as permissive or nonpermissive host reservoirs, respectively. In this review, we highlight the metabolic requirements of Leishmania amastigotes and the evidence that these parasites induce and/or exploit metabolic reprogramming of macrophage metabolism. We also focus on recent studies highlighting the role of key macrophage metabolic signaling pathways, such as mechanistic target of rapamycin, adenosine monophosphate-activated protein kinase and peroxisome proliferator receptor gamma in regulating the pathological progression of Leishmania granulomas. These studies highlight the intimate connectivity between Leishmania and host cell metabolism, the need to investigate these interactions in vivo and the potential to exploit host cell metabolic signaling pathways in developing new host-directed therapies.
Collapse
Affiliation(s)
- Eleanor C Saunders
- Department of Biochemistry and Molecular Biology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Malcolm J McConville
- Department of Biochemistry and Molecular Biology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, VIC, 3010, Australia
| |
Collapse
|
8
|
Silva RCMC, Travassos LH, Paiva CN, Bozza MT. Heme oxygenase-1 in protozoan infections: A tale of resistance and disease tolerance. PLoS Pathog 2020; 16:e1008599. [PMID: 32692767 PMCID: PMC7373268 DOI: 10.1371/journal.ppat.1008599] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Heme oxygenase (HO-1) mediates the enzymatic cleavage of heme, a molecule with proinflammatory and prooxidant properties. HO-1 activity deeply impacts host capacity to tolerate infection through reduction of tissue damage or affecting resistance, the ability of the host to control pathogen loads. In this Review, we will discuss the contribution of HO-1 in different and complex protozoan infections, such as malaria, leishmaniasis, Chagas disease, and toxoplasmosis. The complexity of these infections and the pleiotropic effects of HO-1 constitute an interesting area of study and an opportunity for drug development.
Collapse
Affiliation(s)
- Rafael C. M. C. Silva
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Leonardo H. Travassos
- Laboratório de Imunoreceptores e Sinalização, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudia N. Paiva
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Marcelo T. Bozza
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- * E-mail:
| |
Collapse
|
9
|
Resolvin D1 Administration Is Beneficial in Trypanosoma cruzi Infection. Infect Immun 2020; 88:IAI.00052-20. [PMID: 32152197 DOI: 10.1128/iai.00052-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 03/02/2020] [Indexed: 12/14/2022] Open
Abstract
Chagas disease is a major public health issue, affecting ∼10 million people worldwide. Transmitted by a protozoan named Trypanosoma cruzi, this infection triggers a chronic inflammatory process that can lead to cardiomyopathy (Chagas disease). Resolvin D1 (RvD1) is a novel proresolution lipid mediator whose effects on inflammatory diseases dampens pathological inflammatory responses and can restore tissue homeostasis. Current therapies are not effective in altering the outcome of T. cruzi infection, and as RvD1 has been evaluated as a therapeutic agent in various inflammatory diseases, we examined if exogenous RvD1 could modulate the pathogenesis of Chagas disease in a murine model. CD-1 mice infected with the T. cruzi Brazil strain were treated with RvD1. Mice were administered 3 μg/kg of body weight RvD1 intraperitoneally on days 5, 10, and 15 to examine the effect of RvD1 on acute disease or administered the same dose on days 60, 65, and 70 to examine its effects on chronic infection. RvD1 therapy increased the survival rate and controlled parasite replication in mice with acute infection and reduced the levels of interferon gamma and transforming growth factor β (TGF-β) in mice with chronic infection. In addition, there was an increase in interleukin-10 levels with RvD1 therapy in both mice with acute infection and mice with chronic infection and a decrease in TGF-β levels and collagen content in cardiac tissue. Together, these data indicate that RvD1 therapy can dampen the inflammatory response, promote the resolution of T. cruzi infection, and prevent cardiac fibrosis.
Collapse
|
10
|
Clostridium difficile caused changes in fatty acids profile and resolvin D1 content in plasma of infected patients. Eur J Gastroenterol Hepatol 2020; 32:318-324. [PMID: 31714390 DOI: 10.1097/meg.0000000000001600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Clostridium difficile infection (CDI) is an acute gastrointestinal infection caused by anaerobic, toxin-producing bacteria. During the course of CDI, there is a general inflammatory state. In order to gain a deeper understanding of the role of fatty acids (FAs) in the pathogenesis of acute infection we analyzed their plasma content in both patients with CDI and controls. METHODS The study groups included 40 patients with CDI and 40 healthy volunteers. Plasma FA content was analyzed by gas chromatography, resolvin D1 (RvD1) level using ELISA assay, and we assessed the white blood cell (WBC) count, neutrophil count and C-reactive protein (CRP) level. RESULTS Patients with CDI were characterized by significantly higher values of WBC, neutrophils, platelets and CRP compared with the control group. The saturated FA index was statistically higher and total n-3 FA was significantly decreased in the plasma of CDI patients as compared with the control group. RvD1 content was significantly higher in the control group as compared with patients with CDI. CONCLUSION In patients with good outcomes, we probably observed the effective resolution of inflammation, as reflected in n-3 FA metabolism and their significant decrease in plasma. This may indicate the therapeutic role of n-3 FA in CDI infection.
Collapse
|
11
|
Seyed N, Rafati S, Department of Immunotherapy and leishmania vaccine research, Department of Immunotherapy and Leishmania vaccine research. Resolution and pro-resolving lipid mediators in Leishmania infection. JOURNAL OF MEDICAL MICROBIOLOGY AND INFECTIOUS DISEASES 2019. [DOI: 10.29252/jommid.7.3.61] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
12
|
Paloque L, Perez-Berezo T, Abot A, Dalloux-Chioccioli J, Bourgeade-Delmas S, Le Faouder P, Pujo J, Teste MA, François JM, Schebb NH, Mainka M, Rolland C, Blanpied C, Dietrich G, Bertrand-Michel J, Deraison C, Valentin A, Cenac N. Polyunsaturated fatty acid metabolites: biosynthesis in Leishmania and role in parasite/host interaction. J Lipid Res 2019; 60:636-647. [PMID: 30626624 DOI: 10.1194/jlr.m091736] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/08/2019] [Indexed: 12/20/2022] Open
Abstract
Inside the human host, Leishmania infection starts with phagocytosis of infective promastigotes by macrophages. In order to survive, Leishmania has developed several strategies to manipulate macrophage functions. Among these strategies, Leishmania as a source of bioactive lipids has been poorly explored. Herein, we assessed the biosynthesis of polyunsaturated fatty acid metabolites by infective and noninfective stages of Leishmania and further explored the role of these metabolites in macrophage polarization. The concentration of docosahexaenoic acid metabolites, precursors of proresolving lipid mediators, was increased in the infective stage of the parasite compared with the noninfective stage, and cytochrome P450-like proteins were shown to be implicated in the biosynthesis of these metabolites. The treatment of macrophages with lipids extracted from the infective forms of the parasite led to M2 macrophage polarization and blocked the differentiation into the M1 phenotype induced by IFN-γ. In conclusion, Leishmania polyunsaturated fatty acid metabolites, produced by cytochrome P450-like protein activity, are implicated in parasite/host interactions by promoting the polarization of macrophages into a proresolving M2 phenotype.
Collapse
Affiliation(s)
- Lucie Paloque
- UMR152 Pharma Dev, Université de Toulouse, IRD, UPS, 31400 Toulouse, France.,LCC CNRS, UPR8241, Université de Toulouse, UPS, INPT, 31400 Toulouse, France
| | - Teresa Perez-Berezo
- IRSD, Université de Toulouse, INSERM, INRA, INP-ENVT, 31024 Toulouse, France
| | - Anne Abot
- IRSD, Université de Toulouse, INSERM, INRA, INP-ENVT, 31024 Toulouse, France
| | | | | | | | - Julien Pujo
- IRSD, Université de Toulouse, INSERM, INRA, INP-ENVT, 31024 Toulouse, France
| | - Marie-Ange Teste
- LISBP Université de Toulouse, CNRS, INRA, INSA, 31400 Toulouse, France
| | | | - Nils Helge Schebb
- Faculty of Mathematics and Natural Sciences University of Wuppertal, 42119 Wuppertal, Germany
| | - Malwina Mainka
- Faculty of Mathematics and Natural Sciences University of Wuppertal, 42119 Wuppertal, Germany
| | - Corinne Rolland
- IRSD, Université de Toulouse, INSERM, INRA, INP-ENVT, 31024 Toulouse, France
| | - Catherine Blanpied
- IRSD, Université de Toulouse, INSERM, INRA, INP-ENVT, 31024 Toulouse, France
| | - Gilles Dietrich
- IRSD, Université de Toulouse, INSERM, INRA, INP-ENVT, 31024 Toulouse, France
| | | | - Céline Deraison
- IRSD, Université de Toulouse, INSERM, INRA, INP-ENVT, 31024 Toulouse, France
| | - Alexis Valentin
- UMR152 Pharma Dev, Université de Toulouse, IRD, UPS, 31400 Toulouse, France
| | - Nicolas Cenac
- IRSD, Université de Toulouse, INSERM, INRA, INP-ENVT, 31024 Toulouse, France
| |
Collapse
|
13
|
Luz NF, DeSouza-Vieira T, De Castro W, Vivarini AC, Pereira L, França RR, Silveira-Mattos PS, Costa DL, Teixeira C, Meneses C, Boaventura VS, de Oliveira CI, Lopes UG, Aronson N, Andrade BB, Brodskyn CI, Valenzuela JG, Kamhawi S, Borges VM. Lutzomyia longipalpis Saliva Induces Heme Oxygenase-1 Expression at Bite Sites. Front Immunol 2018; 9:2779. [PMID: 30546363 PMCID: PMC6279893 DOI: 10.3389/fimmu.2018.02779] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 11/12/2018] [Indexed: 12/21/2022] Open
Abstract
Sand flies bite mammalian hosts to obtain a blood meal, driving changes in the host inflammatory response that support the establishment of Leishmania infection. This effect is partially attributed to components of sand fly saliva, which are able to recruit and activate leukocytes. Our group has shown that heme oxygenase-1 (HO-1) favors Leishmania survival in infected cells by reducing inflammatory responses. Here, we show that exposure to sand fly bites is associated with induction of HO-1 in vivo. Histopathological analyses of skin specimens from human volunteers experimentally exposed to sand fly bites revealed that HO-1 and Nrf2 are produced at bite sites in the skin. These results were recapitulated in mice ears injected with a salivary gland sonicate (SGS) or exposed to sand fly bites, indicating that vector saliva may be a key factor in triggering HO-1 expression. Resident skin macrophages were the main source HO-1 at 24–48 h after bites. Additionally, assays in vivo after bites and in vitro after stimulation with saliva both demonstrated that HO-1 production by macrophages was Nrf2-dependent. Collectively, our data demonstrates that vector saliva induces early HO-1 production at the bite sites, representing a major event associated with establishment of naturally-transmitted Leishmania infections.
Collapse
Affiliation(s)
- Nivea F Luz
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Brazil
| | - Thiago DeSouza-Vieira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Waldione De Castro
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Aislan Carvalho Vivarini
- Laboratory of Molecular Parasitology, Center of Health Science, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lais Pereira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Riam Rocha França
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Brazil.,Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
| | - Paulo S Silveira-Mattos
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Brazil.,Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
| | - Diego L Costa
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | | | - Claudio Meneses
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Viviane S Boaventura
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Brazil.,Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
| | - Camila I de Oliveira
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Brazil.,Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
| | - Ulisses Gazos Lopes
- Laboratory of Molecular Parasitology, Center of Health Science, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Naomi Aronson
- Infectious Diseases Division, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Bruno B Andrade
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Brazil.,Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Fundação José Silveira, Salvador, Brazil
| | - Claudia I Brodskyn
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Brazil.,Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
| | - Jesus G Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Valeria M Borges
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Brazil.,Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
| |
Collapse
|
14
|
López-Muñoz RA, Molina-Berríos A, Campos-Estrada C, Abarca-Sanhueza P, Urrutia-Llancaqueo L, Peña-Espinoza M, Maya JD. Inflammatory and Pro-resolving Lipids in Trypanosomatid Infections: A Key to Understanding Parasite Control. Front Microbiol 2018; 9:1961. [PMID: 30186271 PMCID: PMC6113562 DOI: 10.3389/fmicb.2018.01961] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 08/02/2018] [Indexed: 12/30/2022] Open
Abstract
Pathogenic trypanosomatids (Trypanosoma cruzi, Trypanosoma brucei, and Leishmania spp.) are protozoan parasites that cause neglected diseases affecting millions of people in Africa, Asia, and the Americas. In the process of infection, trypanosomatids evade and survive the immune system attack, which can lead to a chronic inflammatory state that induces cumulative damage, often killing the host in the long term. The immune mediators involved in this process are not entirely understood. Most of the research on the immunologic control of protozoan infections has been focused on acute inflammation. Nevertheless, when this process is not terminated adequately, permanent damage to the inflamed tissue may ensue. Recently, a second process, called resolution of inflammation, has been proposed to be a pivotal process in the control of parasite burden and establishment of chronic infection. Resolution of inflammation is an active process that promotes the normal function of injured or infected tissues. Several mediators are involved in this process, including eicosanoid-derived lipids, cytokines such as transforming growth factor (TGF)-β and interleukin (IL)-10, and other proteins such as Annexin-V. For example, during T. cruzi infection, pro-resolving lipids such as 15-epi-lipoxin-A4 and Resolvin D1 have been associated with a decrease in the inflammatory changes observed in experimental chronic heart disease, reducing inflammation and fibrosis, and increasing host survival. Furthermore, Resolvin D1 modulates the immune response in cells of patients with Chagas disease. In Leishmania spp. infections, pro-resolving mediators such as Annexin-V, lipoxins, and Resolvin D1 are related to the modulation of cutaneous manifestation of the disease. However, these mediators seem to have different roles in visceral or cutaneous leishmaniasis. Finally, although T. brucei infections are less well studied in terms of their relationship with inflammation, it has been found that arachidonic acid-derived lipids act as key regulators of the host immune response and parasite burden. Also, cytokines such as IL-10 and TGF-β may be related to increased infection. Knowledge about the inflammation resolution process is necessary to understand the host–parasite interplay, but it also offers an interesting opportunity to improve the current therapies, aiming to reduce the detrimental state induced by chronic protozoan infections.
Collapse
Affiliation(s)
- Rodrigo A López-Muñoz
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Alfredo Molina-Berríos
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Carolina Campos-Estrada
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso, Chile.,Centro de Investigación Farmacopea Chilena, Universidad de Valparaíso, Valparaíso, Chile
| | - Patricio Abarca-Sanhueza
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Luis Urrutia-Llancaqueo
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Miguel Peña-Espinoza
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Juan D Maya
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
15
|
Lima JB, Araújo-Santos T, Lázaro-Souza M, Carneiro AB, Ibraim IC, Jesus-Santos FH, Luz NF, Pontes SDM, Entringer PF, Descoteaux A, Bozza PT, Soares RP, Borges VM. Leishmania infantum lipophosphoglycan induced-Prostaglandin E 2 production in association with PPAR-γ expression via activation of Toll like receptors-1 and 2. Sci Rep 2017; 7:14321. [PMID: 29084985 PMCID: PMC5662570 DOI: 10.1038/s41598-017-14229-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 10/05/2017] [Indexed: 01/16/2023] Open
Abstract
Lipophosphoglycan (LPG) is a key virulence factor expressed on the surfaces of Leishmania promastigotes. Although LPG is known to activate macrophages, the underlying mechanisms resulting in the production of prostaglandin E2 (PGE2) via signaling pathways remain unknown. Here, the inflammatory response arising from stimulation by Leishmania infantum LPG and/or its lipid and glycan motifs was evaluated with regard to PGE2 induction. Intact LPG, but not its glycan and lipid moieties, induced a range of proinflammatory responses, including PGE2 and nitric oxide (NO) release, increased lipid droplet formation, and iNOS and COX2 expression. LPG also induced ERK-1/2 and JNK phosphorylation in macrophages, in addition to the release of PGE2, MCP-1, IL-6, TNF-α and IL-12p70, but not IL-10. Pharmacological inhibition of ERK1/2 and PKC affected PGE2 and cytokine production. Moreover, treatment with rosiglitazone, an agonist of peroxisome proliferator-activated receptor gamma (PPAR-γ), also modulated the release of PGE2 and other proinflammatory mediators. Finally, we determined that LPG-induced PPAR-γ signaling occurred via TLR1/2. Taken together, these results reinforce the role played by L. infantum-derived LPG in the proinflammatory response seen in Leishmania infection.
Collapse
Affiliation(s)
- Jonilson Berlink Lima
- Gonçalo Moniz Institut, Oswaldo Cruz Foundation (FIOCRUZ-BA), 40296-710, Salvador, BA, Brazil.,Center of Biological Sciences and Health, Federal University of Western Bahia (UFOB), 47808-021, Barreiras, BA, Brazil
| | - Théo Araújo-Santos
- Gonçalo Moniz Institut, Oswaldo Cruz Foundation (FIOCRUZ-BA), 40296-710, Salvador, BA, Brazil.,Center of Biological Sciences and Health, Federal University of Western Bahia (UFOB), 47808-021, Barreiras, BA, Brazil
| | - Milena Lázaro-Souza
- Gonçalo Moniz Institut, Oswaldo Cruz Foundation (FIOCRUZ-BA), 40296-710, Salvador, BA, Brazil.,Federal University of Bahia (UFBA), 40110-170, Salvador, BA, Brazil
| | - Alan Brito Carneiro
- Laboratory of Immunopharmacology, Oswaldo Cruz Institut, FIOCRUZ-RJ, 21040-900, Rio de Janeiro, RJ, Brazil
| | - Izabela Coimbra Ibraim
- René Rachou Institut, Oswaldo Cruz Foundation (FIOCRUZ-MG), 30190-002, Belo Horizonte, MG, Brazil
| | - Flávio Henrique Jesus-Santos
- Gonçalo Moniz Institut, Oswaldo Cruz Foundation (FIOCRUZ-BA), 40296-710, Salvador, BA, Brazil.,Federal University of Bahia (UFBA), 40110-170, Salvador, BA, Brazil
| | - Nívea Farias Luz
- Gonçalo Moniz Institut, Oswaldo Cruz Foundation (FIOCRUZ-BA), 40296-710, Salvador, BA, Brazil
| | - Sara de Moura Pontes
- Gonçalo Moniz Institut, Oswaldo Cruz Foundation (FIOCRUZ-BA), 40296-710, Salvador, BA, Brazil.,Federal University of Bahia (UFBA), 40110-170, Salvador, BA, Brazil
| | - Petter Franco Entringer
- Federal University of Rio de Janeiro (UFRJ), NUPEM, Campus Macaé, 27933-378, Macaé, RJ, Brazil
| | - Albert Descoteaux
- Institut National de la Recherche Scientifique, Institut Armand-Frappier, H7V 1B7, Laval, Canada
| | - Patrícia Torres Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institut, FIOCRUZ-RJ, 21040-900, Rio de Janeiro, RJ, Brazil
| | - Rodrigo Pedro Soares
- René Rachou Institut, Oswaldo Cruz Foundation (FIOCRUZ-MG), 30190-002, Belo Horizonte, MG, Brazil.
| | - Valéria Matos Borges
- Gonçalo Moniz Institut, Oswaldo Cruz Foundation (FIOCRUZ-BA), 40296-710, Salvador, BA, Brazil. .,Federal University of Bahia (UFBA), 40110-170, Salvador, BA, Brazil.
| |
Collapse
|
16
|
Anti-parasite therapy drives changes in human visceral leishmaniasis-associated inflammatory balance. Sci Rep 2017; 7:4334. [PMID: 28659627 PMCID: PMC5489532 DOI: 10.1038/s41598-017-04595-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 05/17/2017] [Indexed: 12/29/2022] Open
Abstract
Visceral leishmaniasis (VL) remains a major public health problem worldwide. Cytokine balance is thought to play a critical role in the development of this disease. Here, we perform a prospective exploratory study addressing whether simultaneous assessment of circulating levels of different lipid mediators and cytokines could highlight specific pathways involved with VL pathogenesis. VL patients displayed substantial increases in serum levels of Prostaglandin F2α (PGF2α), Leukotriene B4 (LTB4), Resolvin D1 (RvD1), IL-1β, IL-6, IL-8, IL-10, IL-12p70 and TNF-α compared with uninfected endemic control group, while exhibiting decreased levels of TGF-β1. Hierarchical cluster analysis of the prospective changes in the expression level of theses parameters upon anti-Leishmania treatment initiation revealed that the inflammatory profile observed in active disease gradually changed over time and was generally reversed at day 30 of therapy. Furthermore, not only the individual concentrations of most of the inflammatory biomarkers changed upon treatment, but the correlations between those and several biochemical parameters used to characterize VL disease activity were also modified over time. These results demonstrate that an inflammatory imbalance hallmarks active VL disease and open perspective for manipulation of these pathways in future studies examining a potential host-directed therapy against VL.
Collapse
|