Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Rodrigo Saar Gomes is active.

Publication


Featured researches published by Rodrigo Saar Gomes.


PLOS Neglected Tropical Diseases | 2012

Ecto-nucleotidase activities of promastigotes from leishmania (Viannia) braziliensis relates to parasite infectivity and disease clinical outcome.

Pauline M. Leite; Rodrigo Saar Gomes; Amanda Braga Figueiredo; Tiago Donatelli Serafim; Wagner Luiz Tafuri; Carolina Carvalho de Souza; Sandra Aparecida de Lima Moura; Juliana Lopes Rangel Fietto; Maria Norma Melo; Fátima Ribeiro-Dias; Milton Adriano Pelli de Oliveira; Ana Rabello; Luís Carlos Crocco Afonso

Background Leishmania (Viannia) braziliensis has been associated with a broad range of clinical manifestations ranging from a simple cutaneous ulcer to destructive mucosal lesions. Factors leading to this diversity of clinical presentations are not clear, but parasite factors have lately been recognized as important in determining disease progression. Given the fact that the activity of ecto-nucleotidases correlates with parasitism and the development of infection, we evaluated the activity of these enzymes in promastigotes from 23 L. braziliensis isolates as a possible parasite-related factor that could influence the clinical outcome of the disease. Methodology/Principal Findings Our results show that the isolates differ in their ability to hydrolyze adenine nucleotides. Furthermore, we observed a positive correlation between the time for peak of lesion development in C57BL/6J mice and enzymatic activity and clinical manifestation of the isolate. In addition, we found that L. (V.) braziliensis isolates obtained from mucosal lesions hydrolyze higher amounts of adenine nucleotides than isolates obtained from skin lesions. One isolate with high (PPS6m) and another with low (SSF) ecto-nucleotidase activity were chosen for further studies. Mice inoculated with PPS6m show delayed lesion development and present larger parasite loads than animals inoculated with the SSF isolate. In addition, PPS6m modulates the host immune response by inhibiting dendritic cell activation and NO production by activated J774 macrophages. Finally, we observed that the amastigote forms from PPS6m and SSF isolates present low enzymatic activity that does not interfere with NO production and parasite survival in macrophages. Conclusions/Significance Our data suggest that ecto-nucleotidases present on the promastigote forms of the parasite may interfere with the establishment of the immune response with consequent impaired ability to control parasite dissemination and this may be an important factor in determining the clinical outcome of leishmaniasis.


Journal of Leukocyte Biology | 2017

Interleukin 32: a novel player in the control of infectious diseases

Fátima Ribeiro-Dias; Rodrigo Saar Gomes; Lucas Luiz de Lima Silva; Jéssica Cristina dos Santos; Leo A. B. Joosten

Interleukin 32 (IL‐32) is a proinflammatory cytokine, expressed as 9 distinct isoforms. The most active isoform is the predominantly intracellular‐functioning IL‐32γ. Involvement of IL‐32 in infectious diseases is increasingly being appreciated. Production of IL‐32 promotes pathways that serve to control bacterial infection, especially those caused by mycobacteria. A similar role for this cytokine is observed in the cellular response to viral infections. In addition to its protective effects against microorganisms, IL‐32 is involved in immunopathogenesis of some infectious diseases. In parasitic diseases, it has been demonstrated that this cytokine is induced by Leishmania infection. In this review, we summarize the present data on the role of IL‐32 in infectious diseases, highlighting this cytokine as new target for control of infections.


PLOS Neglected Tropical Diseases | 2017

Cytokines and microbicidal molecules regulated by IL-32 in THP-1-derived human macrophages infected with New World Leishmania species

J.C. Dos Santos; Bas Heinhuis; Rodrigo Saar Gomes; M.S.M.A. Damen; Fernando Real; Renato A. Mortara; S.T. Keating; Charles A. Dinarello; Leo A. B. Joosten; Fátima Ribeiro-Dias

Background Interleukin-32 (IL-32) is expressed in lesions of patients with American Tegumentary Leishmaniasis (ATL), but its precise role in the disease remains unknown. Methodology/Principal findings In the present study, silencing and overexpression of IL-32 was performed in THP-1-derived macrophages infected with Leishmania (Viannia) braziliensis or L. (Leishmania) amazonensis to investigate the role of IL-32 in infection. We report that Leishmania species induces IL-32γ, and show that intracellular IL-32γ protein production is dependent on endogenous TNFα. Silencing or overexpression of IL-32 demonstrated that this cytokine is closely related to TNFα and IL-8. Remarkably, the infection index was augmented in the absence of IL-32 and decreased in cells overexpressing this cytokine. Mechanistically, these effects can be explained by nitric oxide cathelicidin and β-defensin 2 production regulated by IL-32. Conclusions Thus, endogenous IL-32 is a crucial cytokine involved in the host defense against Leishmania parasites.


Cytokine | 2016

Leishmania (Viannia) braziliensis amastigotes induces the expression of TNFα and IL-10 by human peripheral blood mononuclear cells in vitro in a TLR4-dependent manner

Hélio Galdino; Rodrigo Saar Gomes; Jéssica Cristina dos Santos; Lívia Lara Pessoni; Anetícia Eduarda Maldaner; Stefanne Madalena Marques; Clayson Moura Gomes; Miriam Leandro Dorta; Milton Adriano Pelli de Oliveira; Leo A. B. Joosten; Fátima Ribeiro-Dias

While the role of Toll-like receptors (TLRs) has been investigated in murine models of tegumentary leishmaniasis caused by Leishmania (Viannia) braziliensis, the interaction between TLRs and Leishmania sp. has not been investigated in human cells. The aim of this study was to evaluate the involvement of TLR4 in cytokine production of human peripheral blood mononuclear cells (PBMCs) induced by L. braziliensis, and whether the parasite alters the expression of TLR4 on monocytes/macrophages. Amastigote forms were obtained from mice lesions and PBMCs were isolated from healthy donors. PBMCs were cultured in absence or presence of IFNγ, TLR4 neutralizing antibodies, natural antagonist of TLR4 (Bartonella LPS), TLR4 agonist (E. coli LPS), and amastigote forms. The concentrations of tumor necrosis factor (TNFα) and interleukin 10 (IL-10) were assayed by ELISA and TLR4 expression by flow cytometry. Amastigotes forms of L. braziliensis induced TNFα and IL-10 production only in IFNγ-primed PBMCs. The TNFα and IL-10 production was inhibited by TLR4 neutralization, both with anti-TLR4 antibodies and Bartonella LPS. Interestingly, addition of E. coli LPS further increased TNFα but not IL-10 production induced by L. braziliensis amastigotes. Amastigotes of L. braziliensis strongly reduced membrane TLR4 expression on monocytes/macrophages, apparently by internalization after the infection. The present study reveals that TLR4 drives the production of TNFα and IL-10 induced by L. braziliensis amastigotes and that the parasites decrease TLR4 expression on monocyte surface.


Parasites & Vectors | 2017

IL-32γ promotes the healing of murine cutaneous lesions caused by Leishmania braziliensis infection in contrast to Leishmania amazonensis

Rodrigo Saar Gomes; Muriel Vilela Teodoro Silva; Jéssica Cristina dos Santos; Lucas Luiz de Lima Silva; Aline Carvalho Batista; Juliana Reis Machado; Mauro M. Teixeira; Miriam Leandro Dorta; Milton Adriano Pelli de Oliveira; Charles A. Dinarello; Leo A. B. Joosten; Fátima Ribeiro-Dias

BackgroundInterleukin 32 (IL-32) is a pro-inflammatory cytokine induced in patients with American tegumentary leishmaniasis (ATL) caused by Leishmania braziliensis. Here, we investigated whether IL-32 is also expressed in patient lesions caused by L. amazonensis. In addition, we evaluated experimental L. amazonensis and L. braziliensis infections in C57BL/6 transgenic mice for human IL-32γ (IL-32γTg) in comparison with wild-type (WT) mice that do not express the IL-32 gene.ResultsHuman cutaneous lesions caused by L. amazonensis express higher levels of IL-32 than healthy control skin. In mice, the presence of IL-32γ promoted the control of cutaneous lesions caused by L. braziliensis, but not lesions caused by L. amazonensis in an ear dermis infection model. In addition, IL-32γTg mice displayed less tissue parasitism and inflammation in IL-32γTg than WT mice during the healing phase of L. braziliensis infection. Production of antigen-specific pro-inflammatory cytokines was higher in IL-32γTg mice than in WT mice during L. braziliensis infection but not during L. amazonensis infection.ConclusionsHuman cutaneous lesions caused by L. amazonensis express high levels of IL-32. In mice, the presence of IL-32γ contributes to the lesion healing caused by L. braziliensis but not by L. amazonensis. Data suggest that despite the ability for both species to induce IL-32 in humans, the connections between this cytokine and other immune players induced by related species of parasites can lead to distinct outcomes of the murine infections.


Infection and Immunity | 2018

Human Interleukin-32 gamma Plays a Protective Role in an Experimental Model of Visceral Leishmaniasis in Mice

Rodrigo Saar Gomes; Muriel Vilela Teodoro Silva; Jéssica Cristina dos Santos; Christine van Linge; Juliana Machado Reis; Mauro M. Teixeira; Sebastião Alves Pinto; Miriam Leandro Dorta; Xiyuan Bai; Edward D. Chan; Charles A. Dinarello; Milton Adriano Pelli de Oliveira; Leo A. B. Joosten; Fátima Ribeiro-Dias

ABSTRACT Visceral leishmaniasis (VL) is a chronic parasitic disease caused by Leishmania infantum in the Americas. During VL, several proinflammatory cytokines are produced in spleen, liver, and bone marrow. However, the role of interleukin-32 (IL-32) has not been explored in this disease. IL-32 can induce production of proinflammatory cytokines in innate immune cells and polarize the adaptive immune response. Herein, we discovered that L. infantum antigens induced expression of mRNA mainly for the IL-32γ isoform but also induced low levels of the IL-32β transcript in human peripheral blood mononuclear cells. Furthermore, infection of human IL-32γ transgenic mice (IL-32γTg mice) with L. infantum promastigote forms increased IL-32γ expression in the spleen and liver. Interestingly, IL-32γTg mice harbored less parasitism in the spleen and liver than wild-type (WT) mice. In addition, IL-32γTg mice showed increased granuloma formation in the liver compared to WT mice. The protection against VL was associated with increased production of nitric oxide (NO), interferon gamma (IFN-γ), IL-17A, and tumor necrosis factor alpha by splenic cells restimulated ex vivo with L. infantum antigens. In parallel, there was an increase in the number of Th1 and Th17 T cells in the spleens of IL-32γTg mice infected with L. infantum. IL-32γ induction of IFN-γ and IL-17A expression was found to be essential for NO production by splenic cells of infected animals. These data indicate that IL-32γ potentiates the Th1/Th17 immune response during experimental VL, thus contributing to the control of L. infantum infection.


Fems Immunology and Medical Microbiology | 2018

Leishmania (Viannia) guyanensis in tegumentary leishmaniasis

Arissa Felipe Borges; Rodrigo Saar Gomes; Fátima Ribeiro-Dias

Abstract Leishmania (Viannia) guyanensis is a causal agent of American tegumentary leishmaniasis (ATL). This protozoan has been poorly investigated; however, it can cause different clinical forms of ATL, ranging from a single cutaneous lesion to severe lesions that can lead to destruction of the nasopharyngeal mucosa. L. (V.) guyanensis and the disease caused by this species can present unique aspects revealing the need to better characterize this parasite species to improve our knowledge of the immunopathological mechanisms and treatment options for ATL. The mechanisms by which some patients develop a more severe form of ATL remain unclear. It is known that the host immune profile and parasite factors may influence the clinical manifestations of the disease. Besides intrinsic parasite factors, Leishmaniavirus RNA 1 (LRV1) infecting L. guyanensis can contribute to ATL immunopathogenesis. In this review, general aspects of L. guyanensis infection in humans and mouse models are presented.


Scientific Reports | 2017

The NOD2 receptor is crucial for immune responses towards New World Leishmania species

J.C. Dos Santos; M.S.M.A. Damen; Marije Oosting; D.J. de Jong; Bas Heinhuis; Rodrigo Saar Gomes; Mihai G. Netea; Fátima Ribeiro-Dias; Leo A. B. Joosten

American Tegumentary Leishmaniasis is a chronic infection caused by Leishmania protozoan. It is not known whether genetic variances in NOD-like receptor (NLR) family members influence the immune response towards Leishmania parasites and modulate intracellular killing. Using functional genomics, we investigated whether genetic variants in NOD1 or NOD2 influence the production of cytokines by human PBMCs exposed to Leishmania. In addition, we examined whether recognition of Leishmania by NOD2 contributes to intracellular killing. Polymorphisms in the NOD2 gene decreased monocyte- and lymphocyte-derived cytokine production after stimulation with L. amazonensis or L. braziliensis compared to individuals with a functional NOD2 receptor. The phagolysosome formation is important for Leishmania-induced cytokine production and upregulation of NOD2 mRNA expression. NOD2 is crucial to control intracellular infection caused by Leishmania spp. NOD2 receptor is important for Leishmania recognition, the control of intracellular killing, and the induction of innate and adaptive immune responses.


Fems Immunology and Medical Microbiology | 2017

Platelet-activating factor increases reactive oxygen species-mediated microbicidal activity of human macrophages infected with Leishmania (Viannia) braziliensis

Arissa Felipe Borges; Camila Imai Morato; Rodrigo Saar Gomes; Miriam L. Dorta; Milton Adriano Pelli de Oliveira; Fátima Ribeiro-Dias

&NA; Platelet‐activating factor (PAF) is produced by macrophages during inflammation and infections. We evaluated whether PAF is able to modulate the infection of human macrophages by Leishmania braziliensis, the main Leishmania sp. in Brazil. Monocyte‐derived macrophages were incubated with promastigote forms in absence or presence of exogenous PAF. We observed that the treatment of macrophages with low concentrations of PAF prior to infection increased the phagocytosis of L. braziliensis. More importantly, exogenous PAF reduced the parasitism when it was added before, during or after infection. In addition, treatment with a PAF antagonist (PCA 4248) resulted in a significant increase of macrophage infection in a concentration‐dependent manner, suggesting that endogenous PAF is important to control L. braziliensis infection. Mechanistically, while exogenous PAF increased production of reactive oxygen species (ROS) treatment with PCA 4248 reduced oxidative burst during L. braziliensis infection. The microbicidal effects of exogenous PAF were abolished when macrophages were treated with apocynin, an NADPH oxidase inhibitor. The data show that PAF promotes the production of ROS induced by L. braziliensis, suggesting that this lipid mediator may be relevant to control L. braziliensis infection in human macrophages. Graphical Abstract Figure. Figure. This study demonstrates that endogenous or exogenous platelet‐activating factor (PAF) increases induction of reactive oxygen species that are needed to kill Leishmania braziliensis in human macrophages.


Molecules | 2015

The Antileishmanial Potential of C-3 Functionalized Isobenzofuranones against Leishmania (Leishmania) Infantum Chagasi

Wagner Luiz Pereira; Raphael de Souza Vasconcellos; Christiane Mariotini-Moura; Rodrigo Saar Gomes; Rafaela de Cássia Firmino; Adalberto Manoel da Silva; Abelardo Silva Júnior; Gustavo Costa Bressan; Márcia Rogéria de Almeida; Luís Carlos Crocco Afonso; Róbson Ricardo Teixeira; Juliana Lopes Rangel Fietto

Leishmaniases are diseases caused by protozoan parasites of the genus Leishmania. Clinically, leishmaniases range from cutaneous to visceral forms, with estimated global incidences of 1.2 and 0.4 million cases per year, respectively. The treatment of these diseases relies on multiple parenteral injections with pentavalent antimonials or amphotericin B. However, these pharmaceuticals are either too toxic or expensive for routine use in developing countries. These facts call for safer, cheaper, and more effective new antileishmanial drugs. In this investigation, we describe the results of the assessment of the activities of a series of isobenzofuran-1(3H)-ones (phtalides) against Leishmania (Leishmania) infantum chagasi, which is the main causative agent of visceral leishmaniasis in the New World. The compounds were tested at concentrations of 100, 75, 50, 25 and 6.25 µM over 24, 48, and 72 h. After 48 h of treatment at the 100 µM concentration, compounds 7 and 8 decreased parasite viability to 4% and 6%, respectively. The concentration that gives half-maximal responses (LC50) for the antileishmanial activities of compounds 7 and 8 against promastigotes after 24 h were 60.48 and 65.93 µM, respectively. Additionally, compounds 7 and 8 significantly reduced parasite infection in macrophages.Leishmaniases are diseases caused by protozoan parasites of the genus Leishmania. Clinically, leishmaniases range from cutaneous to visceral forms, with estimated global incidences of 1.2 and 0.4 million cases per year, respectively. The treatment of these diseases relies on multiple parenteral injections with pentavalent antimonials or amphotericin B. However, these pharmaceuticals are either too toxic or expensive for routine use in developing countries. These facts call for safer, cheaper, and more effective new antileishmanial drugs. In this investigation, we describe the results of the assessment of the activities of a series of isobenzofuran-1(3H)-ones (phtalides) against Leishmania (Leishmania) infantum chagasi, which is the main causative agent of visceral leishmaniasis in the New World. The compounds were tested at concentrations of 100, 75, 50, 25 and 6.25 µM over 24, 48, and 72 h. After 48 h of treatment at the 100 µM concentration, compounds 7 and 8 decreased parasite viability to 4% and 6%, respectively. The concentration that gives half-maximal responses (LC50) for the antileishmanial activities of compounds 7 and 8 against promastigotes after 24 h were 60.48 and 65.93 µM, respectively. Additionally, compounds 7 and 8 significantly reduced parasite infection in macrophages.

Collaboration


Dive into the Rodrigo Saar Gomes's collaboration.

Top Co-Authors

Avatar

Fátima Ribeiro-Dias

Universidade Federal de Goiás

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Leo A. B. Joosten

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Miriam Leandro Dorta

Universidade Federal de Goiás

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Charles A. Dinarello

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Arissa Felipe Borges

Universidade Federal de Goiás

View shared research outputs
Top Co-Authors

Avatar

Clayson Moura Gomes

Universidade Federal de Goiás

View shared research outputs
Top Co-Authors

Avatar

J.C. Dos Santos

Universidade Federal de Goiás

View shared research outputs
Top Co-Authors

Avatar

Juliana Lopes Rangel Fietto

Universidade Federal de Ouro Preto

View shared research outputs
Researchain Logo
Decentralizing Knowledge