1
|
Oliveira Soté W, Comar Junior M. Exploring Binding Sites in Chagas Disease Protein TcP21 Using Integrated Mixed Solvent Molecular Dynamics Approaches. J Chem Inf Model 2025; 65:363-377. [PMID: 39686861 PMCID: PMC11733930 DOI: 10.1021/acs.jcim.4c01927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 12/18/2024]
Abstract
Chagas disease, caused by the protozoan Trypanosoma cruzi, remains a significant global health burden, particularly in Latin America, where millions are at risk. This disease predominantly affects socioeconomically vulnerable populations, aggravating economic inequality, marginalization, and low political visibility. Despite extensive research, effective treatments are still lacking, partly due to the complex biology of the parasite and its infection mechanisms. This study focuses on TcP21, a novel 21 kDa protein secreted by extracellular amastigotes, which has been implicated in T. cruzi infection via an alternative infective pathway. Although the potential of TcP21 for understanding Chagas disease is promising, further exploration is necessary, particularly in identifying potential binding sites on its surface. Computational tools offer a versatile and effective strategy for preliminary binding site assessment, facilitating a more cost-efficient allocation of experimental resources. In this study, we employed three independent computational approaches─mixed solvent molecular dynamics simulations (MSMD), fragment-based molecular docking, and pharmacophore model docking coupled with molecular dynamics simulations─to identify potential binding sites and provide comprehensive insights into TcP21. The three methodologies converged on a common site located on the external surface of the protein, characterized by key residues such as GLU55, ASP52, VAL70, ILE62, and TRP77. The protonated amino, acetamido, and phenyl groups of the pharmacophore probe were consistently observed to interact with the site via a network of salt bridges, hydrogen bonds, charge-charge interactions, and alkyl-π interactions, suggesting these groups play a significant role in ligand binding. This study does not aim to propose specific therapeutic hits but to highlight a still unknown and unexplored protein involved in T. cruzi cell invasion. In this regard, given the strong correlation between the three distinct approaches used for mapping, we consider this study offers valuable insights for further research into P21 and its role in Chagas disease.
Collapse
Affiliation(s)
- William Oliveira Soté
- Institute of Chemistry, Federal University of Uberlândia, Uberlândia 38400-902, Brazil
| | - Moacyr Comar Junior
- Institute of Chemistry, Federal University of Uberlândia, Uberlândia 38400-902, Brazil
| |
Collapse
|
2
|
Teixeira TL, Teixeira SC, Borges BC, Servato JPS, Oliveira ECMD, Velikkakam T, Silva CVD. Trypanosoma cruzi P21 protein exacerbates Leishmania (L.) amazonensis infection. Exp Parasitol 2024; 267:108856. [PMID: 39461608 DOI: 10.1016/j.exppara.2024.108856] [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: 03/26/2024] [Revised: 09/26/2024] [Accepted: 10/24/2024] [Indexed: 10/29/2024]
Abstract
The protozoan parasite Trypanosoma cruzi, the etiological agent of Chagas disease, affects millions of people worldwide. Current treatments rely on drugs effective only in the acute phase, making the search for new therapeutic targets a priority. While a recombinant protein based on T. cruzi P21 (rP21) exhibits immunomodulatory properties and contributes to controlling parasitism and inflammation during T. cruzi infection, its efficacy against other trypanosomatids remains unexplored. This study investigated the impact of rP21 on Leishmania (L.) amazonensis infection in a murine model. Contrary to our expectations, treatment with rP21 did not ameliorate L. (L.) amazonensis infection. Instead, rP21 treatment resulted in increased parasite load in the paws of infected BALB/c mice, evidenced by larger lesion sizes and higher parasite burdens, accompanied by an intensified inflammatory infiltrate in the paw tissue. These findings suggest that despite its promising effects in the context of T. cruzi infection, rP21 may not be a suitable therapeutic candidate for L. amazonensis infection and might even exacerbate disease.
Collapse
Affiliation(s)
- Thaise Lara Teixeira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Samuel Cota Teixeira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Bruna Cristina Borges
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | | | | | - Teresiama Velikkakam
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Claudio Vieira da Silva
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil.
| |
Collapse
|
3
|
Silveira ACA, Uombe NPI, Velikkakam T, Borges BC, Teixeira TL, de Almeida VF, Torres JDA, Pereira CL, de Souza G, Teixeira SC, Servato JPS, Silva MJB, Mineo TWP, Ribas RM, Mortara RA, da Silveira JF, da Silva CV. The Trypanosoma cruzi pleiotropic protein P21 orchestrates the intracellular retention and in-vivo parasitism control of virulent Y strain parasites. Front Cell Infect Microbiol 2024; 14:1412345. [PMID: 38988814 PMCID: PMC11233463 DOI: 10.3389/fcimb.2024.1412345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/11/2024] [Indexed: 07/12/2024] Open
Abstract
P21 is a protein secreted by all forms of Trypanosoma cruzi (T. cruzi) with recognized biological activities determined in studies using the recombinant form of the protein. In our recent study, we found that the ablation of P21 gene decreased Y strain axenic epimastigotes multiplication and increased intracellular replication of amastigotes in HeLa cells infected with metacyclic trypomastigotes. In the present study, we investigated the effect of P21 in vitro using C2C12 cell lines infected with tissue culture-derived trypomastigotes (TCT) of wild-type and P21 knockout (TcP21-/-) Y strain, and in vivo using an experimental model of T. cruzi infection in BALB/c mice. Our in-vitro results showed a significant decrease in the host cell invasion rate by TcP21-/- parasites as measured by Giemsa staining and cell count in bright light microscope. Quantitative polymerase chain reaction (qPCR) analysis showed that TcP21-/- parasites multiplied intracellularly to a higher extent than the scrambled parasites at 72h post-infection. In addition, we observed a higher egress of TcP21-/- trypomastigotes from C2C12 cells at 144h and 168h post-infection. Mice infected with Y strain TcP21-/- trypomastigotes displayed higher systemic parasitemia, heart tissue parasite burden, and several histopathological alterations in heart tissues compared to control animals infected with scrambled parasites. Therewith, we propose that P21 is important in the host-pathogen interaction during invasion, cell multiplication, and egress, and may be part of the mechanism that controls parasitism and promotes chronic infection without patent systemic parasitemia.
Collapse
Affiliation(s)
| | - Nelsa Paula Inácio Uombe
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Teresiama Velikkakam
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Bruna Cristina Borges
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Thaise Lara Teixeira
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | - Cecília Luiza Pereira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Guilherme de Souza
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Samuel Cota Teixeira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | | | | | | | - Rosineide Marques Ribas
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Renato Arruda Mortara
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - José Franco da Silveira
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Claudio Vieira da Silva
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| |
Collapse
|
4
|
de Souza G, Teixeira SC, Fajardo Martínez AF, Silva RJ, Luz LC, de Lima Júnior JP, Rosini AM, dos Santos NCL, de Oliveira RM, Paschoalino M, Barbosa MC, Alves RN, Gomes AO, da Silva CV, Ferro EAV, Barbosa BF. Trypanosoma cruzi P21 recombinant protein modulates Toxoplasma gondii infection in different experimental models of the human maternal-fetal interface. Front Immunol 2023; 14:1243480. [PMID: 37915581 PMCID: PMC10617204 DOI: 10.3389/fimmu.2023.1243480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/25/2023] [Indexed: 11/03/2023] Open
Abstract
Introduction Toxoplasma gondii is the etiologic agent of toxoplasmosis, a disease that affects about one-third of the human population. Most infected individuals are asymptomatic, but severe cases can occur such as in congenital transmission, which can be aggravated in individuals infected with other pathogens, such as HIV-positive pregnant women. However, it is unknown whether infection by other pathogens, such as Trypanosoma cruzi, the etiologic agent of Chagas disease, as well as one of its proteins, P21, could aggravate T. gondii infection. Methods In this sense, we aimed to investigate the impact of T. cruzi and recombinant P21 (rP21) on T. gondii infection in BeWo cells and human placental explants. Results Our results showed that T. cruzi infection, as well as rP21, increases invasion and decreases intracellular proliferation of T. gondii in BeWo cells. The increase in invasion promoted by rP21 is dependent on its binding to CXCR4 and the actin cytoskeleton polymerization, while the decrease in proliferation is due to an arrest in the S/M phase in the parasite cell cycle, as well as interleukin (IL)-6 upregulation and IL-8 downmodulation. On the other hand, in human placental villi, rP21 can either increase or decrease T. gondii proliferation, whereas T. cruzi infection increases T. gondii proliferation. This increase can be explained by the induction of an anti-inflammatory environment through an increase in IL-4 and a decrease in IL-6, IL-8, macrophage migration inhibitory factor (MIF), and tumor necrosis factor (TNF)-α production. Discussion In conclusion, in situations of coinfection, the presence of T. cruzi may favor the congenital transmission of T. gondii, highlighting the importance of neonatal screening for both diseases, as well as the importance of studies with P21 as a future therapeutic target for the treatment of Chagas disease, since it can also favor T. gondii infection.
Collapse
Affiliation(s)
- Guilherme de Souza
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Samuel Cota Teixeira
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Aryani Felixa Fajardo Martínez
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Rafaela José Silva
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Luana Carvalho Luz
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Joed Pires de Lima Júnior
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Alessandra Monteiro Rosini
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Natália Carine Lima dos Santos
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Rafael Martins de Oliveira
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Marina Paschoalino
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Matheus Carvalho Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Rosiane Nascimento Alves
- Department of Agricultural and Natural Science, Universidade do Estado de Minas Gerais, Ituiutaba, MG, Brazil
| | - Angelica Oliveira Gomes
- Institute of Natural and Biological Sciences, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil
| | - Claudio Vieira da Silva
- Laboratory of Trypanosomatids, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Eloisa Amália Vieira Ferro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Bellisa Freitas Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| |
Collapse
|
5
|
Azevedo Silveira AC, Cristina de Oliveira R, Rodrigues CC, Teixeira SC, Borges BC, Vieira da Silva C. Trypanosoma cruzi infection induces proliferation and impairs migration of a human breast cancer cell line. Exp Parasitol 2023; 245:108443. [PMID: 36526003 DOI: 10.1016/j.exppara.2022.108443] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/10/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022]
Abstract
Breast cancer is considered the type of cancer that most affects women in the world. The triple negative breast cancer is considered aggressive with poor prognosis. In the 1930s Russian researchers observed that T. cruzi has tropism for tumor cells. Since then, this research field has been subject of a numerous of researches. Here, we proposed to investigate the impact of T. cruzi infection on proliferation and migration of triple negative breast cancer cell line (MDA-MB-231). T. cruzi showed high invasion and multiplication rate in MDA-MB-231 cell line. The infection promoted the multiplication of MDA-MB-231 cell, continuous cell lysis throughout of days of in vitro infection and impaired MDA-MB-231 cell migration. Taken together, these results demonstrated the high susceptibility of MDA-MB-231 cell to T. cruzi and suggested that molecules from T. cruzi may impair host cell migration with potential use to avoid metastasis.
Collapse
Affiliation(s)
| | | | - Cassiano Costa Rodrigues
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Samuel Cota Teixeira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Bruna Cristina Borges
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil.
| | - Claudio Vieira da Silva
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil.
| |
Collapse
|
6
|
Oliveira MM, Bonturi CR, Salu BR, Oliva MLV, Mortara RA, Orikaza CM. Modulation of STAT-1, STAT-3, and STAT-6 activities in THP-1 derived macrophages infected with two Trypanosoma cruzi strains. Front Immunol 2022; 13:1038332. [PMID: 36389843 PMCID: PMC9643828 DOI: 10.3389/fimmu.2022.1038332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/04/2022] [Indexed: 12/03/2022] Open
Abstract
Trypanosoma cruzi is the causative protozoan of Chagas' Disease, a neglected tropical disease that affects 6-7 million people worldwide. Interaction of the parasite with the host immune system is a key factor in disease progression and chronic symptoms. Although the human immune system is capable of controlling the disease, the parasite has numerous evasion mechanisms that aim to maintain intracellular persistence and survival. Due to the pronounced genetic variability of T. cruzi, co-infections or mixed infections with more than one parasite strain have been reported in the literature. The intermodulation in such cases is unclear. This study aimed to evaluate the co-infection of T. cruzi strains G and CL compared to their individual infections in human macrophages derived from THP-1 cells activated by classical or alternative pathways. Flow cytometry analysis demonstrated that trypomastigotes were more infective than extracellular amastigotes (EAs) and that strain G could infect more macrophages than strain CL. Classically activated macrophages showed lower number of infected cells and IL-4-stimulated cells displayed increased CL-infected macrophages. However, co-infection was a rare event. CL EAs decreased the production of reactive oxygen species (ROS), whereas G trypomastigotes displayed increased ROS detection in classically activated cells. Co-infection did not affect ROS production. Monoinfection by strain G or CL mainly induced an anti-inflammatory cytokine profile by decreasing inflammatory cytokines (IFN-γ, TNF-α, IL-1β) and/or increasing IL-4, IL-10, and TGF-β. Co-infection led to a predominant inflammatory milieu, with reduced IL-10 and TGF-β, and/or promotion of IFN-γ and IL-1β release. Infection by strain G reduced activation of intracellular signal transducer and activator of transcription (STAT) factors. In EAs, monoinfections impaired STAT-1 activity and promoted phosphorylation of STAT-3, both changes may prolong cell survival. Coinfected macrophages displayed pronounced activation of all STATs examined. These activations likely promoted parasite persistence and survival of infected cells. The collective results demonstrate that although macrophages respond to both strains, T. cruzi can modulate the intracellular environment, inducing different responses depending on the strain, parasite infective form, and co-infection or monoinfection. The modulation influences parasite persistence and survival of infected cells.
Collapse
Affiliation(s)
- Melissa Martins Oliveira
- ¹Microbiology, Immunology and Parasitology Department, Escola Paulista de Medicina, Federal University of São Paulo - UNIFESP, São Paulo, Brazil
| | - Camila Ramalho Bonturi
- ²Biochemistry Department, Escola Paulista de Medicina, Federal University of São Paulo - UNIFESP, São Paulo, Brazil
| | - Bruno Ramos Salu
- ²Biochemistry Department, Escola Paulista de Medicina, Federal University of São Paulo - UNIFESP, São Paulo, Brazil
| | - Maria Luiza Vilela Oliva
- ²Biochemistry Department, Escola Paulista de Medicina, Federal University of São Paulo - UNIFESP, São Paulo, Brazil
| | - Renato Arruda Mortara
- ¹Microbiology, Immunology and Parasitology Department, Escola Paulista de Medicina, Federal University of São Paulo - UNIFESP, São Paulo, Brazil
| | - Cristina Mary Orikaza
- ¹Microbiology, Immunology and Parasitology Department, Escola Paulista de Medicina, Federal University of São Paulo - UNIFESP, São Paulo, Brazil
| |
Collapse
|
7
|
Umarao P, Rath PP, Gourinath S. Cdc42/Rac Interactive Binding Containing Effector Proteins in Unicellular Protozoans With Reference to Human Host: Locks of the Rho Signaling. Front Genet 2022; 13:781885. [PMID: 35186026 PMCID: PMC8847673 DOI: 10.3389/fgene.2022.781885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/14/2022] [Indexed: 11/23/2022] Open
Abstract
Small GTPases are the key to actin cytoskeleton signaling, which opens the lock of effector proteins to forward the signal downstream in several cellular pathways. Actin cytoskeleton assembly is associated with cell polarity, adhesion, movement and other functions in eukaryotic cells. Rho proteins, specifically Cdc42 and Rac, are the primary regulators of actin cytoskeleton dynamics in higher and lower eukaryotes. Effector proteins, present in an inactive state gets activated after binding to the GTP bound Cdc42/Rac to relay a signal downstream. Cdc42/Rac interactive binding (CRIB) motif is an essential conserved sequence found in effector proteins to interact with Cdc42 or Rac. A diverse range of Cdc42/Rac and their effector proteins have evolved from lower to higher eukaryotes. The present study has identified and further classified CRIB containing effector proteins in lower eukaryotes, focusing on parasitic protozoans causing neglected tropical diseases and taking human proteins as a reference point to the highest evolved organism in the evolutionary trait. Lower eukaryotes’ CRIB containing proteins fall into conventional effector molecules, PAKs (p21 activated kinase), Wiskoit-Aldrich Syndrome proteins family, and some have unique domain combinations unlike any known proteins. We also highlight the correlation between the effector protein isoforms and their selective specificity for Cdc42 or Rac proteins during evolution. Here, we report CRIB containing effector proteins; ten in Dictyostelium and Entamoeba, fourteen in Acanthamoeba, one in Trypanosoma and Giardia. CRIB containing effector proteins that have been studied so far in humans are potential candidates for drug targets in cancer, neurological disorders, and others. Conventional CRIB containing proteins from protozoan parasites remain largely elusive and our data provides their identification and classification for further in-depth functional validations. The tropical diseases caused by protozoan parasites lack combinatorial drug targets as effective paradigms. Targeting signaling mechanisms operative in these pathogens can provide greater molecules in combatting their infections.
Collapse
Affiliation(s)
- Preeti Umarao
- Structural Biology Lab, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Pragyan Parimita Rath
- Structural Biology Lab, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Samudrala Gourinath
- Structural Biology Lab, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
8
|
Teixeira TL, Chiurillo MA, Lander N, Rodrigues CC, Onofre TS, Ferreira ÉR, Yonamine CM, Santos JDG, Mortara RA, da Silva CV, da Silveira JF. Ablation of the P21 Gene of Trypanosoma cruzi Provides Evidence of P21 as a Mediator in the Control of Epimastigote and Intracellular Amastigote Replication. Front Cell Infect Microbiol 2022; 12:799668. [PMID: 35252026 PMCID: PMC8895596 DOI: 10.3389/fcimb.2022.799668] [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: 10/21/2021] [Accepted: 01/25/2022] [Indexed: 11/23/2022] Open
Abstract
P21 is an immunomodulatory protein expressed throughout the life cycle of Trypanosoma cruzi, the etiologic agent of Chagas disease. In vitro and in vivo studies have shown that P21 plays an important role in the invasion of mammalian host cells and establishment of infection in a murine model. P21 functions as a signal transducer, triggering intracellular cascades in host cells and resulting in the remodeling of the actin cytoskeleton and parasite internalization. Furthermore, in vivo studies have shown that P21 inhibits angiogenesis, induces inflammation and fibrosis, and regulates intracellular amastigote replication. In this study, we used the CRISPR/Cas9 system for P21 gene knockout and investigated whether the ablation of P21 results in changes in the phenotypes associated with this protein. Ablation of P21 gene resulted in a lower growth rate of epimastigotes and delayed cell cycle progression, accompanied by accumulation of parasites in G1 phase. However, P21 knockout epimastigotes were viable and able to differentiate into metacyclic trypomastigotes, which are infective to mammalian cells. In comparison with wild-type parasites, P21 knockout cells showed a reduced cell invasion rate, demonstrating the role of this protein in host cell invasion. However, there was a higher number of intracellular amastigotes per cell, suggesting that P21 is a negative regulator of amastigote proliferation in mammalian cells. Here, for the first time, we demonstrated the direct correlation between P21 and the replication of intracellular amastigotes, which underlies the chronicity of T. cruzi infection.
Collapse
Affiliation(s)
- Thaise Lara Teixeira
- Departmento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Miguel Angel Chiurillo
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, United States
| | - Noelia Lander
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, United States
| | | | - Thiago Souza Onofre
- Departmento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Éden Ramalho Ferreira
- Departmento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Camila Miyagui Yonamine
- Departmento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Renato Arruda Mortara
- Departmento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Claudio Vieira da Silva
- Laboratório de Tripanosomatídeos, Universidade Federal de Uberlândia, Uberlândia, Brazil
- *Correspondence: Claudio Vieira da Silva, ; José Franco da Silveira,
| | - José Franco da Silveira
- Departmento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
- *Correspondence: Claudio Vieira da Silva, ; José Franco da Silveira,
| |
Collapse
|
9
|
Dick CF, Rocco-Machado N, Dos-Santos ALA, Carvalho-Kelly LF, Alcantara CL, Cunha-E-Silva NL, Meyer-Fernandes JR, Vieyra A. An Iron Transporter Is Involved in Iron Homeostasis, Energy Metabolism, Oxidative Stress, and Metacyclogenesis in Trypanosoma cruzi. Front Cell Infect Microbiol 2022; 11:789401. [PMID: 35083166 PMCID: PMC8785980 DOI: 10.3389/fcimb.2021.789401] [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: 10/04/2021] [Accepted: 12/06/2021] [Indexed: 11/29/2022] Open
Abstract
The parasite Trypanosoma cruzi causes Chagas’ disease; both heme and ionic Fe are required for its optimal growth, differentiation, and invasion. Fe is an essential cofactor in many metabolic pathways. Fe is also harmful due to catalyzing the formation of reactive O2 species; for this reason, all living systems develop mechanisms to control the uptake, metabolism, and storage of Fe. However, there is limited information available on Fe uptake by T. cruzi. Here, we identified a putative 39-kDa Fe transporter in T. cruzi genome, TcIT, homologous to the Fe transporter in Leishmania amazonensis and Arabidopsis thaliana. Epimastigotes grown in Fe-depleted medium have increased TcIT transcription compared with controls grown in regular medium. Intracellular Fe concentration in cells maintained in Fe-depleted medium is lower than in controls, and there is a lower O2 consumption. Epimastigotes overexpressing TcIT, which was encountered in the parasite plasma membrane, have high intracellular Fe content, high O2 consumption—especially in phosphorylating conditions, high intracellular ATP, very high H2O2 production, and stimulated transition to trypomastigotes. The investigation of the mechanisms of Fe transport at the cellular and molecular levels will assist in elucidating Fe metabolism in T. cruzi and the involvement of its transport in the differentiation from epimastigotes to trypomastigotes, virulence, and maintenance/progression of the infection.
Collapse
Affiliation(s)
- Claudia F Dick
- Leopoldo de Meis Institute of Medical Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Center of Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nathália Rocco-Machado
- Leopoldo de Meis Institute of Medical Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Center of Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - André L A Dos-Santos
- Leopoldo de Meis Institute of Medical Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Center of Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luiz F Carvalho-Kelly
- Leopoldo de Meis Institute of Medical Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Center of Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carolina L Alcantara
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Center of Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Narcisa L Cunha-E-Silva
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Center of Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - José R Meyer-Fernandes
- Leopoldo de Meis Institute of Medical Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Center of Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Adalberto Vieyra
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Center of Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Graduate Program in Translational Biomedicine/BIOTRANS, Unigranrio University, Duque de Caxias, Brazil
| |
Collapse
|
10
|
Rodríguez-Bejarano OH, Avendaño C, Patarroyo MA. Mechanisms Associated with Trypanosoma cruzi Host Target Cell Adhesion, Recognition and Internalization. Life (Basel) 2021; 11:534. [PMID: 34207491 PMCID: PMC8227291 DOI: 10.3390/life11060534] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/02/2021] [Accepted: 06/04/2021] [Indexed: 12/11/2022] Open
Abstract
Chagas disease is caused by the kinetoplastid parasite Trypanosoma cruzi, which is mainly transmitted by hematophagous insect bites. The parasite's lifecycle has an obligate intracellular phase (amastigotes), while metacyclic and bloodstream-trypomastigotes are its infective forms. Mammalian host cell recognition of the parasite involves the interaction of numerous parasite and host cell plasma membrane molecules and domains (known as lipid rafts), thereby ensuring internalization by activating endocytosis mechanisms triggered by various signaling cascades in both host cells and the parasite. This increases cytoplasmatic Ca2+ and cAMP levels; cytoskeleton remodeling and endosome and lysosome intracellular system association are triggered, leading to parasitophorous vacuole formation. Its membrane becomes modified by containing the parasite's infectious form within it. Once it has become internalized, the parasite seeks parasitophorous vacuole lysis for continuing its intracellular lifecycle, fragmenting such a vacuole's membrane. This review covers the cellular and molecular mechanisms involved in T. cruzi adhesion to, recognition of and internalization in host target cells.
Collapse
Affiliation(s)
- Oscar Hernán Rodríguez-Bejarano
- Health Sciences Faculty, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Calle 222#55-37, Bogotá 111166, Colombia;
| | - Catalina Avendaño
- Animal Science Faculty, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Calle 222#55-37, Bogotá 111166, Colombia;
| | - Manuel Alfonso Patarroyo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50#26-20, Bogotá 111321, Colombia
- Health Sciences Division, Main Campus, Universidad Santo Tomás, Carrera 9#51-11, Bogotá 110231, Colombia
- Microbiology Department, Faculty of Medicine, Universidad Nacional de Colombia, Carrera 45#26-85, Bogotá 111321, Colombia
| |
Collapse
|
11
|
de Castro Neto AL, da Silveira JF, Mortara RA. Comparative Analysis of Virulence Mechanisms of Trypanosomatids Pathogenic to Humans. Front Cell Infect Microbiol 2021; 11:669079. [PMID: 33937106 PMCID: PMC8085324 DOI: 10.3389/fcimb.2021.669079] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/30/2021] [Indexed: 11/23/2022] Open
Abstract
Trypanosoma brucei, Leishmania spp., and T. cruzi are flagellate protozoans of the family Trypanosomatidae and the causative agents of human African trypanosomiasis, leishmaniasis, and Chagas disease, respectively. These diseases affect humans worldwide and exert a significant impact on public health. Over the course of evolution, the parasites associated with these pathologies have developed mechanisms to circumvent the immune response system throughout the infection cycle. In cases of human infection, this function is undertaken by a group of proteins and processes that allow the parasites to propagate and survive during host invasion. In T. brucei, antigenic variation is promoted by variant surface glycoproteins and other proteins involved in evasion from the humoral immune response, which helps the parasite sustain itself in the extracellular milieu during infection. Conversely, Leishmania spp. and T. cruzi possess a more complex infection cycle, with specific intracellular stages. In addition to mechanisms for evading humoral immunity, the pathogens have also developed mechanisms for facilitating their adhesion and incorporation into host cells. In this review, the different immune evasion strategies at cellular and molecular levels developed by these human-pathogenic trypanosomatids have been discussed, with a focus on the key molecules responsible for mediating the invasion and evasion mechanisms and the effects of these molecules on virulence.
Collapse
Affiliation(s)
- Artur Leonel de Castro Neto
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - José Franco da Silveira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Renato Arruda Mortara
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
12
|
Ferri G, Edreira MM. All Roads Lead to Cytosol: Trypanosoma cruzi Multi-Strategic Approach to Invasion. Front Cell Infect Microbiol 2021; 11:634793. [PMID: 33747982 PMCID: PMC7973469 DOI: 10.3389/fcimb.2021.634793] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/27/2021] [Indexed: 12/17/2022] Open
Abstract
T. cruzi has a complex life cycle involving four developmental stages namely, epimastigotes, metacyclic trypomastigotes, amastigotes and bloodstream trypomastigotes. Although trypomastigotes are the infective forms, extracellular amastigotes have also shown the ability to invade host cells. Both stages can invade a broad spectrum of host tissues, in fact, almost any nucleated cell can be the target of infection. To add complexity, the parasite presents high genetic variability with differential characteristics such as infectivity. In this review, we address the several strategies T. cruzi has developed to subvert the host cell signaling machinery in order to gain access to the host cell cytoplasm. Special attention is made to the numerous parasite/host protein interactions and to the set of signaling cascades activated during the formation of a parasite-containing vesicle, the parasitophorous vacuole, from which the parasite escapes to the cytosol, where differentiation and replication take place.
Collapse
Affiliation(s)
- Gabriel Ferri
- CONICET-Universidad de Buenos Aires, IQUIBICEN, Ciudad de Buenos Aires, Argentina
| | - Martin M Edreira
- CONICET-Universidad de Buenos Aires, IQUIBICEN, Ciudad de Buenos Aires, Argentina.,Laboratorio de Biología Molecular de Trypanosoma, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos, Ciudad de Buenos Aires, Argentina.,Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
13
|
Borges BC, Uehara IA, Dos Santos MA, Martins FA, de Souza FC, Junior ÁF, da Luz FAC, da Costa MS, Notário AFO, Lopes DS, Teixeira SC, Teixeira TL, de Castilhos P, da Silva CV, Silva MJB. The Recombinant Protein Based on Trypanosoma cruzi P21 Interacts With CXCR4 Receptor and Abrogates the Invasive Phenotype of Human Breast Cancer Cells. Front Cell Dev Biol 2020; 8:569729. [PMID: 33195200 PMCID: PMC7604327 DOI: 10.3389/fcell.2020.569729] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/22/2020] [Indexed: 01/07/2023] Open
Abstract
Trypanosoma cruzi P21 is a protein secreted by the parasite that plays biological roles directly involved in the progression of Chagas disease. The recombinant protein (rP21) demonstrates biological properties, such as binding to CXCR4 receptors in macrophages, chemotactic activity of immune cells, and inhibiting angiogenesis. This study aimed to verify the effects of rP21 interaction with CXCR4 from non-tumoral cells (MCF-10A) and triple-negative breast cancer cells (MDA-MB-231). Our data showed that the MDA-MB-231 cells expressed higher levels of CXCR4 than did the non-tumor cell lines. Besides, cytotoxicity assays using different concentrations of rP21 showed that the recombinant protein was non-toxic and was able to bind to the cell membranes of both cell lineages. In addition, rP21 reduced the migration and invasion of MDA-MB-231 cells by the downregulation of MMP-9 gene expression. In addition, treatment with rP21 blocked the cell cycle, arresting it in the G1 phase, mainly in MDA-MB-231 cells. Finally, rP21 prevents the chemotaxis and proliferation induced by CXCL12. Our data showed that rP21 binds to the CXCR4 receptors in both cells, downregulates CXCR4 gene expression, and decreases the receptors in the cytoplasm of MDA-MB-231 cells, suggesting CXCR4 internalization. This internalization may explain the desensitization of the receptors in these cells. Thus, rP21 prevents migration, invasion, and progression in MDA-MB-231 cells.
Collapse
Affiliation(s)
- Bruna Cristina Borges
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil.,Laboratório de Biomarcadores Tumorais e Osteoimunologia, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Isadora Akemi Uehara
- Laboratório de Biomarcadores Tumorais e Osteoimunologia, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Marlus Alves Dos Santos
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Flávia Alves Martins
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Fernanda Carvalho de Souza
- Laboratório de Biomarcadores Tumorais e Osteoimunologia, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Álvaro Ferreira Junior
- Departamento de MedicinaVeterinária, Escola de Veterinária e Zootecnia, Universidade Federal de Goiás, Goiânia, Brazil
| | - Felipe Andrés Cordero da Luz
- Laboratório de Biomarcadores Tumorais e Osteoimunologia, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Mylla Spirandelli da Costa
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Ana Flávia Oliveira Notário
- Laboratório de Nanobiotecnologia, Instituto de Genético e Bioquímica, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Daiana Silva Lopes
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Brazil
| | - Samuel Cota Teixeira
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Thaise Lara Teixeira
- Laboratório de Biologia Molecular de Trypanosoma Cruzi, Departamento de Parasitologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Patrícia de Castilhos
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Claudio Vieira da Silva
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Marcelo José Barbosa Silva
- Laboratório de Biomarcadores Tumorais e Osteoimunologia, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| |
Collapse
|
14
|
Martins FA, Dos Santos MA, Santos JDG, da Silva AA, Borges BC, da Costa MS, Tavares PCB, Teixeira SC, Brígido RTES, Teixeira TL, Rodrigues CC, Silva NSDL, de Oliveira RC, de Faria LC, Lemes MR, Zanon RG, Tomiosso TC, Machado JR, da Silva MV, Oliveira CJF, da Silva CV. The Recombinant Form of Trypanosoma cruzi P21 Controls Infection by Modulating Host Immune Response. Front Immunol 2020; 11:1010. [PMID: 32655546 PMCID: PMC7325895 DOI: 10.3389/fimmu.2020.01010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 04/28/2020] [Indexed: 12/22/2022] Open
Abstract
Trypanosoma cruzi P21 protein (P21) is a putative secreted and immunomodulatory molecule with potent bioactive properties such as induction of phagocytosis and actin cytoskeleton polymerization. Despite the bioactive properties described so far, the action of P21 on parasite replication in muscle cell lineage or T. cruzi parasitism during acute experimental infection is unclear. We observed that recombinant P21 (rP21) decreased the multiplication of T. cruzi in C2C12 myoblasts, phenomenon associated with greater actin polymerization and IFN-γ and IL-4 higher expression. During experimental infection, lower cardiac nests, inflammatory infiltrate and fibrosis were observed in mice infected and treated with rP21. These results were correlated with large expression of IFN-γ counterbalanced by high levels of IL-10, which was consistent with the lower cardiac tissue injury found in these mice. We have also observed that upon stress, such as that induced by the presence of the IFN-γ cytokine, T. cruzi produced more P21. The effect of P21 in controlling the replication of T. cruzi, may indicate an evolutionary mechanism of survival developed by the parasite. Thus, when subjected to different stress conditions, the protozoan produces more P21, which induces T. cruzi latency in the host organism, enabling the protozoan to evade the host's immune system.
Collapse
Affiliation(s)
- Flávia Alves Martins
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Marlus Alves Dos Santos
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Júlia de Gouveia Santos
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Aline Alves da Silva
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Bruna Cristina Borges
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Mylla Spirandelli da Costa
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Paula Cristina Brígido Tavares
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Samuel Cota Teixeira
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Rebecca Tavares E Silva Brígido
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Thaise Lara Teixeira
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil.,Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Cassiano Costa Rodrigues
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | | | - Rayane Cristina de Oliveira
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Laura Caroline de Faria
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal do Triângulo Mineiro, Uberaba, Brazil
| | - Marcela Rezende Lemes
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal do Triângulo Mineiro, Uberaba, Brazil
| | - Renata Graciele Zanon
- Departamento de Anatomia Humana, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Tatiana Carla Tomiosso
- Setor de Histologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Juliana Reis Machado
- Departamento de Patologia, Genética e Evolução, Universidade Federal do Triangulo Mineiro, Uberaba, Brazil
| | - Marcos Vinicius da Silva
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal do Triângulo Mineiro, Uberaba, Brazil
| | - Carlo José Freire Oliveira
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal do Triângulo Mineiro, Uberaba, Brazil
| | - Claudio Vieira da Silva
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| |
Collapse
|
15
|
Teixeira TL, Castilhos P, Rodrigues CC, da Silva AA, Brígido RT, Teixeira SC, Borges BC, Dos Santos MA, Martins FA, Santos PCF, Servato JPS, Silva MS, da Silva MJB, Elias MC, da Silva CV. Experimental evidences that P21 protein controls Trypanosoma cruzi replication and modulates the pathogenesis of infection. Microb Pathog 2019; 135:103618. [PMID: 31310832 DOI: 10.1016/j.micpath.2019.103618] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/04/2019] [Accepted: 07/06/2019] [Indexed: 10/26/2022]
Abstract
P21 is a protein secreted by Trypanosoma cruzi (T. cruzi). Previous studies have shown a spectrum of biological activities performed by P21 such as induction of phagocytosis, leukocyte chemotaxis and inhibition of angiogenesis. However, the activity of P21 in T. cruzi infection remains unknown. Here, we reported the role of P21 in mice harboring late T. cruzi infection. Treatment with recombinant P21 protein (rP21) reduced parasite load and angiogenesis, and induced fibrosis in the cardiac tissue of infected mice. In addition, rP21 reduced the growth of epimastigotes, inhibited intracellular replication of amastigotes and modulated the parasite cell cycle. Our data suggest that P21 controls parasite replication in the host, supporting the survival of both parasite and host.
Collapse
Affiliation(s)
- Thaise L Teixeira
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Rua Piauí, Bloco 2B Sala 200 Campus Umuarama, 38400-902, Uberlândia, MG, Brazil
| | - Patrícia Castilhos
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Rua Piauí, Bloco 2B Sala 200 Campus Umuarama, 38400-902, Uberlândia, MG, Brazil
| | - Cassiano C Rodrigues
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Rua Piauí, Bloco 2B Sala 200 Campus Umuarama, 38400-902, Uberlândia, MG, Brazil
| | - Aline A da Silva
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Rua Piauí, Bloco 2B Sala 200 Campus Umuarama, 38400-902, Uberlândia, MG, Brazil
| | - Rebecca Ts Brígido
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Rua Piauí, Bloco 2B Sala 200 Campus Umuarama, 38400-902, Uberlândia, MG, Brazil
| | - Samuel C Teixeira
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Rua Piauí, Bloco 2B Sala 200 Campus Umuarama, 38400-902, Uberlândia, MG, Brazil
| | - Bruna C Borges
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Rua Piauí, Bloco 2B Sala 200 Campus Umuarama, 38400-902, Uberlândia, MG, Brazil
| | - Marlus A Dos Santos
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Rua Piauí, Bloco 2B Sala 200 Campus Umuarama, 38400-902, Uberlândia, MG, Brazil
| | - Flávia A Martins
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Rua Piauí, Bloco 2B Sala 200 Campus Umuarama, 38400-902, Uberlândia, MG, Brazil
| | - Paulo César F Santos
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo - São Paulo, Brazil
| | | | - M S Silva
- Centro de Toxinas, Resposta Imune e Sinalização Celular, Instituto Butantan, São Paulo, São Paulo, Brazil
| | - M J B da Silva
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - M C Elias
- Centro de Toxinas, Resposta Imune e Sinalização Celular, Instituto Butantan, São Paulo, São Paulo, Brazil
| | - Claudio V da Silva
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Rua Piauí, Bloco 2B Sala 200 Campus Umuarama, 38400-902, Uberlândia, MG, Brazil.
| |
Collapse
|
16
|
Pérez Brandán C, Mesias AC, Acuña L, Teixeira TL, da Silva CV. Evaluation of pathogen P21 protein as a potential modulator of the protective immunity induced by Trypanosoma cruzi attenuated parasites. Mem Inst Oswaldo Cruz 2019; 114:e180571. [PMID: 31116244 PMCID: PMC6528380 DOI: 10.1590/0074-02760180571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 04/08/2019] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND TcP21 is a ubiquitous secreted protein of Trypanosoma cruzi and its recombinant form (rP21) promotes parasite cell invasion and acts as a phagocytosis inducer by activating actin polymerisation in the host cell. OBJECTIVE Our goal was to evaluate if the additional supplementation of rP21 during a prime/boost/challenge scheme with T. cruzi TCC attenuated parasites could modify the well-known protective behavior conferred by these parasites. METHODS The humoral immune response was evaluated through the assessment of total anti-T. cruzi antibodies as well as IgG subtypes. IFN-γ, TNF-α and IL-10 were measured in supernatants of splenic cells stimulated with total parasite homogenate or rP21. FINDINGS Our results demonstrated that, when comparing TCC+rP21 vs. TCC vaccinated animals, the levels of IFN-γ were significantly higher in the former group, while the levels of IL-10 and TNF-α were significantly lower. Further, the measurement of parasite load after lethal challenge showed an exacerbated infection and parasite load in heart and skeletal muscle after pre-treatment with rP21, suggesting the important role of this protein during parasite natural invasion process. MAIN CONCLUSION Our results demonstrated that rP21 may have adjuvant capacity able to modify the cytokine immune profile elicited by attenuated parasites.
Collapse
Affiliation(s)
- Cecilia Pérez Brandán
- Universidad Nacional de Salta, Instituto de Patología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Salta- Capital, Argentina
| | - Andrea C Mesias
- Universidad Nacional de Salta, Instituto de Patología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Salta- Capital, Argentina
| | - Leonardo Acuña
- Universidad Nacional de Salta, Instituto de Patología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Salta- Capital, Argentina
| | - Thaise Lara Teixeira
- Universidade Federal de Uberlândia, Instituto de Ciências Biomédicas, Uberlândia, MG, Brasil
| | - Claudio Vieira da Silva
- Universidade Federal de Uberlândia, Instituto de Ciências Biomédicas, Uberlândia, MG, Brasil
| |
Collapse
|
17
|
Bonfim-Melo A, Ferreira ER, Florentino PTV, Mortara RA. Amastigote Synapse: The Tricks of Trypanosoma cruzi Extracellular Amastigotes. Front Microbiol 2018; 9:1341. [PMID: 30013522 PMCID: PMC6036244 DOI: 10.3389/fmicb.2018.01341] [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: 04/04/2018] [Accepted: 05/31/2018] [Indexed: 12/11/2022] Open
Abstract
To complete its life cycle within the mammalian host, Trypanosoma cruzi, the agent of Chagas’ disease, must enter cells. Trypomastigotes originating from the insect vector (metacyclic) or from infected cells (bloodstream/tissue culture-derived) are the classical infective forms of the parasite and enter mammalian cells in an actin-independent manner. By contrast, amastigotes originating from the premature rupture of infected cells or transformed from swimming trypomastigotes (designated extracellular amastigotes, EAs) require functional intact microfilaments to invade non-phagocytic host cells. Earlier work disclosed the key features of EA-HeLa cell interplay: actin-rich protrusions called ‘cups’ are formed at EA invasion sites on the host cell membrane that are also enriched in actin-binding proteins, integrins and extracellular matrix elements. In the past decades we described the participation of membrane components and secreted factors from EAs as well as the actin-regulating proteins of host cells involved in what we propose to be a phagocytic-like mechanism of parasite uptake. Thus, regarding this new perspective herein we present previously described EA-induced ‘cups’ as parasitic synapse since they can play a role beyond its architecture function. In this review, we focus on recent findings that shed light on the intricate interaction between extracellular amastigotes and non-phagocytic HeLa cells.
Collapse
Affiliation(s)
- Alexis Bonfim-Melo
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Eden R Ferreira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Pilar T V Florentino
- DNA Repair Lab, Biomedical Sciences Institute II, Universidade de São Paulo, São Paulo, Brazil
| | - Renato A Mortara
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
18
|
Holzmuller P, Geiger A, Nzoumbou-Boko R, Pissarra J, Hamrouni S, Rodrigues V, Dauchy FA, Lemesre JL, Vincendeau P, Bras-Gonçalves R. Trypanosomatid Infections: How Do Parasites and Their Excreted-Secreted Factors Modulate the Inducible Metabolism of l-Arginine in Macrophages? Front Immunol 2018; 9:778. [PMID: 29731753 PMCID: PMC5921530 DOI: 10.3389/fimmu.2018.00778] [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: 11/12/2017] [Accepted: 03/28/2018] [Indexed: 12/20/2022] Open
Abstract
Mononuclear phagocytes (monocytes, dendritic cells, and macrophages) are among the first host cells to face intra- and extracellular protozoan parasites such as trypanosomatids, and significant expansion of macrophages has been observed in infected hosts. They play essential roles in the outcome of infections caused by trypanosomatids, as they can not only exert a powerful antimicrobial activity but also promote parasite proliferation. These varied functions, linked to their phenotypic and metabolic plasticity, are exerted via distinct activation states, in which l-arginine metabolism plays a pivotal role. Depending on the environmental factors and immune response elements, l-arginine metabolites contribute to parasite elimination, mainly through nitric oxide (NO) synthesis, or to parasite proliferation, through l-ornithine and polyamine production. To survive and adapt to their hosts, parasites such as trypanosomatids developed mechanisms of interaction to modulate macrophage activation in their favor, by manipulating several cellular metabolic pathways. Recent reports emphasize that some excreted-secreted (ES) molecules from parasites and sugar-binding host receptors play a major role in this dialog, particularly in the modulation of the macrophage's inducible l-arginine metabolism. Preventing l-arginine dysregulation by drugs or by immunization against trypanosomatid ES molecules or by blocking partner host molecules may control early infection and is a promising way to tackle neglected diseases including Chagas disease, leishmaniases, and African trypanosomiases. The present review summarizes recent knowledge on trypanosomatids and their ES factors with regard to their influence on macrophage activation pathways, mainly the NO synthase/arginase balance. The review ends with prospects for the use of biological knowledge to develop new strategies of interference in the infectious processes used by trypanosomatids, in particular for the development of vaccines or immunotherapeutic approaches.
Collapse
Affiliation(s)
- Philippe Holzmuller
- CIRAD, Montpellier, France.,UMR 117 ASTRE "Animal, Santé, Territoire, Risques et Ecosystèmes", Univ. Montpellier (I-MUSE), CIRAD, INRA, Montpellier, France
| | - Anne Geiger
- UMR 177 INTERTRYP "Interactions Hôte-Vecteur-Parasite-Environnement dans les maladies tropicales négligées dues aux Trypanosomatidae", Univ. Montpellier (I-MUSE), CIRAD, IRD, Univ. Bordeaux 2, Univ. Lyon 1, Montpellier, France
| | - Romaric Nzoumbou-Boko
- UMR 177 INTERTRYP "Interactions Hôte-Vecteur-Parasite-Environnement dans les maladies tropicales négligées dues aux Trypanosomatidae", Univ. Montpellier (I-MUSE), CIRAD, IRD, Univ. Bordeaux 2, Univ. Lyon 1, Montpellier, France.,Univ. Bordeaux, UMR 177 INTERTRYP, Bordeaux, France.,CHU Bordeaux, Laboratoire de Parasitologie-Mycologie, Bordeaux, France
| | - Joana Pissarra
- UMR 177 INTERTRYP "Interactions Hôte-Vecteur-Parasite-Environnement dans les maladies tropicales négligées dues aux Trypanosomatidae", Univ. Montpellier (I-MUSE), CIRAD, IRD, Univ. Bordeaux 2, Univ. Lyon 1, Montpellier, France
| | - Sarra Hamrouni
- UMR 177 INTERTRYP "Interactions Hôte-Vecteur-Parasite-Environnement dans les maladies tropicales négligées dues aux Trypanosomatidae", Univ. Montpellier (I-MUSE), CIRAD, IRD, Univ. Bordeaux 2, Univ. Lyon 1, Montpellier, France
| | - Valérie Rodrigues
- CIRAD, Montpellier, France.,UMR 117 ASTRE "Animal, Santé, Territoire, Risques et Ecosystèmes", Univ. Montpellier (I-MUSE), CIRAD, INRA, Montpellier, France
| | - Frédéric-Antoine Dauchy
- UMR 177 INTERTRYP "Interactions Hôte-Vecteur-Parasite-Environnement dans les maladies tropicales négligées dues aux Trypanosomatidae", Univ. Montpellier (I-MUSE), CIRAD, IRD, Univ. Bordeaux 2, Univ. Lyon 1, Montpellier, France.,Univ. Bordeaux, UMR 177 INTERTRYP, Bordeaux, France.,CHU Bordeaux, Département des Maladies Infectieuses et Tropicales, Bordeaux, France
| | - Jean-Loup Lemesre
- UMR 177 INTERTRYP "Interactions Hôte-Vecteur-Parasite-Environnement dans les maladies tropicales négligées dues aux Trypanosomatidae", Univ. Montpellier (I-MUSE), CIRAD, IRD, Univ. Bordeaux 2, Univ. Lyon 1, Montpellier, France
| | - Philippe Vincendeau
- UMR 177 INTERTRYP "Interactions Hôte-Vecteur-Parasite-Environnement dans les maladies tropicales négligées dues aux Trypanosomatidae", Univ. Montpellier (I-MUSE), CIRAD, IRD, Univ. Bordeaux 2, Univ. Lyon 1, Montpellier, France.,Univ. Bordeaux, UMR 177 INTERTRYP, Bordeaux, France.,CHU Bordeaux, Laboratoire de Parasitologie-Mycologie, Bordeaux, France
| | - Rachel Bras-Gonçalves
- UMR 177 INTERTRYP "Interactions Hôte-Vecteur-Parasite-Environnement dans les maladies tropicales négligées dues aux Trypanosomatidae", Univ. Montpellier (I-MUSE), CIRAD, IRD, Univ. Bordeaux 2, Univ. Lyon 1, Montpellier, France
| |
Collapse
|
19
|
Florentino PTV, Real F, Orikaza CM, da Cunha JPC, Vitorino FNL, Cordero EM, Sobreira TJP, Mortara RA. A Carbohydrate Moiety of Secreted Stage-Specific Glycoprotein 4 Participates in Host Cell Invasion by Trypanosoma cruzi Extracellular Amastigotes. Front Microbiol 2018; 9:693. [PMID: 29692765 PMCID: PMC5903068 DOI: 10.3389/fmicb.2018.00693] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 03/23/2018] [Indexed: 11/23/2022] Open
Abstract
Trypanosoma cruzi is the etiologic agent of Chagas' disease. It is known that amastigotes derived from trypomastigotes in the extracellular milieu are infective in vitro and in vivo. Extracellular amastigotes (EAs) have a stage-specific surface antigen called Ssp-4, a GPI-anchored glycoprotein that is secreted by the parasites. By immunoprecipitation with the Ssp-4-specific monoclonal antibodies (mAb) 2C2 and 1D9, we isolated the glycoprotein from EAs. By mass spectrometry, we identified the core protein of Ssp-4 and evaluated mRNA expression and the presence of Ssp-4 carbohydrate epitopes recognized by mAb1D9. We demonstrated that the carbohydrate epitope recognized by mAb1D9 could promote host cell invasion by EAs. Although infectious EAs express lower amounts of Ssp-4 compared with less-infectious EAs (at the mRNA and protein levels), it is the glycosylation of Ssp-4 (identified by mAb1D9 staining only in infectious strains and recognized by galectin-3 on host cells) that is the determinant of EA invasion of host cells. Furthermore, Ssp-4 is secreted by EAs, either free or associated with parasite vesicles, and can participate in host-cell interactions. The results presented here describe the possible role of a carbohydrate moiety of T. cruzi surface glycoproteins in host cell invasion by EA forms, highlighting the potential of these moieties as therapeutic and vaccine targets for the treatment of Chagas' disease.
Collapse
Affiliation(s)
- Pilar T. V. Florentino
- Department of Microbiology, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | | | - Cristina M. Orikaza
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Julia P. C. da Cunha
- Special Laboratory of Cell Cycle, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo, Brazil
| | - Francisca N. L. Vitorino
- Special Laboratory of Cell Cycle, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo, Brazil
| | - Esteban M. Cordero
- Department of Microbiology, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
- Facultad de Ciencias, Centro de Genómica y Bioinformática, Universidad Mayor, Santiago, Chile
| | | | - Renato A. Mortara
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
20
|
Borges BC, Uehara IA, Dias LOS, Brígido PC, da Silva CV, Silva MJB. Mechanisms of Infectivity and Evasion Derived from Microvesicles Cargo Produced by Trypanosoma cruzi. Front Cell Infect Microbiol 2016; 6:161. [PMID: 27921011 PMCID: PMC5118865 DOI: 10.3389/fcimb.2016.00161] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/07/2016] [Indexed: 11/13/2022] Open
Abstract
Cell invasion by the intracellular protozoans requires interaction of proteins from both the host and the parasite. Many parasites establish chronic infections, showing they have the potential to escape the immune system; for example, Trypanosoma cruzi is an intracellular parasite that causes Chagas disease. Parasite internalization into host cell requires secreted and surface molecules, such as microvesicles. The release of microvesicles and other vesicles, such as exosomes, by different eukaryotic organisms was first observed in the late twentieth century. The characterization and function of these vesicles have recently been the focus of several investigations. In this review, we discuss the release of microvesicles by T. cruzi. The molecular content of these vesicles is composed of several molecules that take place during parasite-host cell interaction and contribute to the parasite-driven mechanism of evasion from the host immune system. These new findings appear to have a profound impact on the comprehension of T. cruzi biology and highlight novel potential strategies for developing more efficient therapeutic approaches.
Collapse
Affiliation(s)
- Bruna C Borges
- Laboratório de Osteoimunologia e Imunologia dos Tumores, Instituto de Ciências Biomédicas, Universidade Federal de UberlândiaUberlândia, Brazil; Laboratório de Tripanossomatídeos, Instituto de Ciências Biomédicas, Universidade Federal de UberlândiaUberlândia, Brazil
| | - Isadora A Uehara
- Laboratório de Osteoimunologia e Imunologia dos Tumores, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia Uberlândia, Brazil
| | - Laysa O S Dias
- Laboratório de Osteoimunologia e Imunologia dos Tumores, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia Uberlândia, Brazil
| | - Paula C Brígido
- Laboratório de Tripanossomatídeos, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia Uberlândia, Brazil
| | - Claudio V da Silva
- Laboratório de Tripanossomatídeos, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia Uberlândia, Brazil
| | - Marcelo J B Silva
- Laboratório de Osteoimunologia e Imunologia dos Tumores, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia Uberlândia, Brazil
| |
Collapse
|
21
|
Ohyama K, Huy NT, Yoshimi H, Kishikawa N, Nishizawa JE, Roca Y, Revollo Guzmán RJ, Velarde FUG, Kuroda N, Hirayama K. Proteomic profile of circulating immune complexes in chronic Chagas disease. Parasite Immunol 2016; 38:609-17. [PMID: 27223052 DOI: 10.1111/pim.12341] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 04/22/2016] [Indexed: 12/21/2022]
Abstract
Immune complexes (ICs) are the direct and real-time products of humoral immune responses. The identification of constituent foreign or autoantigens within ICs might bring new insights into the pathology of infectious diseases. We applied immune complexome analysis of plasma to the study of Chagas disease caused by Trypanosoma cruzi. Twenty seropositive plasma samples including cardiac and/or megacolon determinate patients (n = 11) and indeterminate (n = 9) were analysed along with 10 seronegative individuals to characterize the antigens bound to circulating ICs. We identified 39 T. cruzi antigens and 114 human autoantigens specific to patients with Chagas. Among those antigens, two T. cruzi antigens (surface protease GP63, glucose-6-isomerase) and six human autoantigens (CD180 antigen, ceruloplasmin, fibrinogen beta chain, fibrinogen beta chain isoform 2 preprotein, isoform gamma-A of fibrinogen γ-chain, serum paraoxonase) were detected in more than 50% of the patients tested. Human isoform short of complement factor H-related protein 2 and trans-sialidase of T. cruzi were more frequently found in the indeterminate (5/9 for both) compared with in the determinate Chagas (0/11, P = 0·046 for human, 1/11, P = 0·0498 for T. cruzi). The immune complexome could illustrate the difference of immune status between clinical forms of chronic Chagas disease.
Collapse
Affiliation(s)
- K Ohyama
- Course of Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.,Nagasaki University Research Centre for Genomic Instability and Carcinogenesis (NRGIC), Nagasaki, Japan
| | - N T Huy
- Department of Clinical Product Development, Institute of Tropical Medicine (NEKKEN), and Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - H Yoshimi
- Course of Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - N Kishikawa
- Course of Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | | | - Y Roca
- Centro Nacional de Enfermedades Tropicales (CENETROP), Santa Cruz, Bolivia
| | - R J Revollo Guzmán
- Centro Nacional de Enfermedades Tropicales (CENETROP), Santa Cruz, Bolivia
| | | | - N Kuroda
- Course of Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.
| | - K Hirayama
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), and Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.
| |
Collapse
|
22
|
Ferreira ÉR, Horjales E, Bonfim-Melo A, Cortez C, da Silva CV, De Groote M, Sobreira TJP, Cruz MC, Lima FM, Cordero EM, Yoshida N, da Silveira JF, Mortara RA, Bahia D. Unique behavior of Trypanosoma cruzi mevalonate kinase: A conserved glycosomal enzyme involved in host cell invasion and signaling. Sci Rep 2016; 6:24610. [PMID: 27113535 PMCID: PMC4845012 DOI: 10.1038/srep24610] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 03/29/2016] [Indexed: 11/30/2022] Open
Abstract
Mevalonate kinase (MVK) is an essential enzyme acting in early steps of sterol isoprenoids biosynthesis, such as cholesterol in humans or ergosterol in trypanosomatids. MVK is conserved from bacteria to mammals, and localizes to glycosomes in trypanosomatids. During the course of T. cruzi MVK characterization, we found that, in addition to glycosomes, this enzyme may be secreted and modulate cell invasion. To evaluate the role of TcMVK in parasite-host cell interactions, TcMVK recombinant protein was produced and anti-TcMVK antibodies were raised in mice. TcMVK protein was detected in the supernatant of cultures of metacyclic trypomastigotes (MTs) and extracellular amastigotes (EAs) by Western blot analysis, confirming its secretion into extracellular medium. Recombinant TcMVK bound in a non-saturable dose-dependent manner to HeLa cells and positively modulated internalization of T. cruzi EAs but inhibited invasion by MTs. In HeLa cells, TcMVK induced phosphorylation of MAPK pathway components and proteins related to actin cytoskeleton modifications. We hypothesized that TcMVK is a bifunctional enzyme that in addition to playing a classical role in isoprenoid synthesis in glycosomes, it is secreted and may modulate host cell signaling required for T. cruzi invasion.
Collapse
Affiliation(s)
- Éden Ramalho Ferreira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | | | - Alexis Bonfim-Melo
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Cristian Cortez
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Claudio Vieira da Silva
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | | | | | - Mário Costa Cruz
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Fabio Mitsuo Lima
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Esteban Mauricio Cordero
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Nobuko Yoshida
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - José Franco da Silveira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Renato Arruda Mortara
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Diana Bahia
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil.,Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, MG, Brazil
| |
Collapse
|
23
|
Watanabe Costa R, da Silveira JF, Bahia D. Interactions between Trypanosoma cruzi Secreted Proteins and Host Cell Signaling Pathways. Front Microbiol 2016; 7:388. [PMID: 27065960 PMCID: PMC4814445 DOI: 10.3389/fmicb.2016.00388] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 03/11/2016] [Indexed: 12/13/2022] Open
Abstract
Chagas disease is one of the prevalent neglected tropical diseases, affecting at least 6-7 million individuals in Latin America. It is caused by the protozoan parasite Trypanosoma cruzi, which is transmitted to vertebrate hosts by blood-sucking insects. After infection, the parasite invades and multiplies in the myocardium, leading to acute myocarditis that kills around 5% of untreated individuals. T. cruzi secretes proteins that manipulate multiple host cell signaling pathways to promote host cell invasion. The primary secreted lysosomal peptidase in T. cruzi is cruzipain, which has been shown to modulate the host immune response. Cruzipain hinders macrophage activation during the early stages of infection by interrupting the NF-kB P65 mediated signaling pathway. This allows the parasite to survive and replicate, and may contribute to the spread of infection in acute Chagas disease. Another secreted protein P21, which is expressed in all of the developmental stages of T. cruzi, has been shown to modulate host phagocytosis signaling pathways. The parasite also secretes soluble factors that exert effects on host extracellular matrix, such as proteolytic degradation of collagens. Finally, secreted phospholipase A from T. cruzi contributes to lipid modifications on host cells and concomitantly activates the PKC signaling pathway. Here, we present a brief review of the interaction between secreted proteins from T. cruzi and the host cells, emphasizing the manipulation of host signaling pathways during invasion.
Collapse
Affiliation(s)
- Renata Watanabe Costa
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo São Paulo, Brazil
| | - Jose F da Silveira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo São Paulo, Brazil
| | - Diana Bahia
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São PauloSão Paulo, Brazil; Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas GeraisMinas Gerais, Brazil
| |
Collapse
|
24
|
Teixeira TL, Machado FC, Alves da Silva A, Teixeira SC, Borges BC, Dos Santos MA, Martins FA, Brígido PC, Rodrigues AA, Notário AFO, Ferreira BA, Servato JPS, Deconte SR, Lopes DS, Ávila VMR, Araújo FDA, Tomiosso TC, Silva MJB, da Silva CV. Trypanosoma cruzi P21: a potential novel target for chagasic cardiomyopathy therapy. Sci Rep 2015; 5:16877. [PMID: 26574156 PMCID: PMC4648062 DOI: 10.1038/srep16877] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 10/21/2015] [Indexed: 11/09/2022] Open
Abstract
Chagas disease, which is caused by the parasite Trypanosoma cruzi, is an important cause of cardiomyopathy in Latin America. It is estimated that 10%–30% of all infected individuals will acquire chronic chagasic cardiomyopathy (CCC). The etiology of CCC is multifactorial and involves parasite genotype, host genetic polymorphisms, immune response, signaling pathways and autoimmune progression. Herein we verified the impact of the recombinant form of P21 (rP21), a secreted T. cruzi protein involved in host cell invasion, on progression of inflammatory process in a polyester sponge-induced inflammation model. Results indicated that rP21 can recruit immune cells induce myeloperoxidase and IL-4 production and decrease blood vessels formation compared to controls in vitro and in vivo. In conclusion, T. cruzi P21 may be a potential target for the development of P21 antagonist compounds to treat chagasic cardiomyopathy.
Collapse
Affiliation(s)
- Thaise Lara Teixeira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, MG, Brasil
| | - Fabrício Castro Machado
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, MG, Brasil.,Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, SP, Brasil
| | - Aline Alves da Silva
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, MG, Brasil
| | - Samuel Cota Teixeira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, MG, Brasil
| | | | | | - Flávia Alves Martins
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, MG, Brasil
| | | | - Adele Aud Rodrigues
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, MG, Brasil
| | | | | | | | - Simone Ramos Deconte
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, MG, Brasil
| | - Daiana Silva Lopes
- Instituto de Genética e Bioquímica, Universidade Federal de Uberlândia, MG, Brasil
| | | | | | | | | | | |
Collapse
|
25
|
A successful strategy for the recovering of active P21, an insoluble recombinant protein of Trypanosoma cruzi. Sci Rep 2014; 4:4259. [PMID: 24590372 PMCID: PMC3941101 DOI: 10.1038/srep04259] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 02/13/2014] [Indexed: 01/26/2023] Open
Abstract
Structural studies of proteins normally require large quantities of pure material that can only be obtained through heterologous expression systems and recombinant technique. In these procedures, large amounts of expressed protein are often found in the insoluble fraction, making protein purification from the soluble fraction inefficient, laborious, and costly. Usually, protein refolding is avoided due to a lack of experimental assays that can validate correct folding and that can compare the conformational population to that of the soluble fraction. Herein, we propose a validation method using simple and rapid 1D 1H nuclear magnetic resonance (NMR) spectra that can efficiently compare protein samples, including individual information of the environment of each proton in the structure.
Collapse
|
26
|
Teixeira TL, Teixeira SC, da Silva CV, de Souza MA. Potential therapeutic use of herbal extracts in trypanosomiasis. Pathog Glob Health 2014; 108:30-6. [PMID: 24548158 PMCID: PMC4083165 DOI: 10.1179/2047773213y.0000000120] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The aim of the present study was to evaluate the effects of crude extracts from Handroanthus impetiginosa, Ageratum conyzoides, and Ruta graveolens on Leishmania amazonensis and Trypanosoma cruzi infection in vitro. The results showed that the extracts caused significant toxicity in promastigotes and trypomastigotes. A significant decrease in the rate of cell invasion by pretreated trypomastigotes and promastigotes was also observed. The extracts caused a significant reduction of the multiplication of intracellular amastigotes of both parasites. Therefore, these herbal extracts may be potential candidates for the development of drugs for the treatment of leishmaniasis and Chagas disease.
Collapse
Affiliation(s)
- Thaise L Teixeira
- Laboratório de Tripanosomatídeos, Instituto de Ciências Biomédicas, Universidade Federal De Uberlândia, Uberlândia, MG, Brazil
| | - Samuel Cota Teixeira
- Laboratório de Tripanosomatídeos, Instituto de Ciências Biomédicas, Universidade Federal De Uberlândia, Uberlândia, MG, Brazil
| | - Claudio Vieira da Silva
- Laboratório de Tripanosomatídeos, Instituto de Ciências Biomédicas, Universidade Federal De Uberlândia, Uberlândia, MG, Brazil
| | - Maria A de Souza
- Laboratório de Tripanosomatídeos, Instituto de Ciências Biomédicas, Universidade Federal De Uberlândia, Uberlândia, MG, Brazil
| |
Collapse
|
27
|
Bernabó G, Levy G, Ziliani M, Caeiro LD, Sánchez DO, Tekiel V. TcTASV-C, a protein family in Trypanosoma cruzi that is predominantly trypomastigote-stage specific and secreted to the medium. PLoS One 2013; 8:e71192. [PMID: 23923058 PMCID: PMC3726618 DOI: 10.1371/journal.pone.0071192] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 06/27/2013] [Indexed: 12/20/2022] Open
Abstract
Among the several multigene families codified by the genome of T. cruzi, the TcTASV family was the latest discovered. The TcTASV (Trypomastigote, Alanine, Serine, Valine) family is composed of ∼40 members, with conserved carboxi- and amino-termini but with a variable central core. According to the length and sequence of the central region the family is split into 3 subfamilies. The TcTASV family is conserved in the genomes of – at least – lineages TcI and TcVI and has no orthologues in other trypanosomatids. In the present work we focus on the study of the TcTASV-C subfamily, composed by 16 genes in the CL Brener strain. We determined that TcTASV-C is preferentially expressed in trypomastigotes, but it is not a major component of the parasite. Both immunoflourescence and flow cytometry experiments indicated that TcTASV-C has a clonal expression, i.e. it is not expressed by all the parasites of a certain population at the same time. We also determined that TcTASV-C is phosphorylated and glycosylated. TASV-C is attached to the parasite surface by a GPI anchor and is shed spontaneously into the medium. About 30% of sera from infected hosts reacted with TcTASV-C, confirming its exposition to the immune system. Its superficial localization and secretory nature suggest a possible role in host-parasite interactions.
Collapse
Affiliation(s)
- Guillermo Bernabó
- Instituto de Investigaciones Biotecnológicas – Instituto Tecnológico de Chascomus (IIB-INTECH), Universidad Nacional de San Martín (UNSAM) – Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Gabriela Levy
- Instituto de Investigaciones Biotecnológicas – Instituto Tecnológico de Chascomus (IIB-INTECH), Universidad Nacional de San Martín (UNSAM) – Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María Ziliani
- Instituto de Investigaciones Biotecnológicas – Instituto Tecnológico de Chascomus (IIB-INTECH), Universidad Nacional de San Martín (UNSAM) – Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Lucas D. Caeiro
- Instituto de Investigaciones Biotecnológicas – Instituto Tecnológico de Chascomus (IIB-INTECH), Universidad Nacional de San Martín (UNSAM) – Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Daniel O. Sánchez
- Instituto de Investigaciones Biotecnológicas – Instituto Tecnológico de Chascomus (IIB-INTECH), Universidad Nacional de San Martín (UNSAM) – Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Valeria Tekiel
- Instituto de Investigaciones Biotecnológicas – Instituto Tecnológico de Chascomus (IIB-INTECH), Universidad Nacional de San Martín (UNSAM) – Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
28
|
Cruz MC, Souza-Melo N, da Silva CV, DaRocha WD, Bahia D, Araújo PR, Teixeira SR, Mortara RA. Trypanosoma cruzi: role of δ-amastin on extracellular amastigote cell invasion and differentiation. PLoS One 2012; 7:e51804. [PMID: 23272170 PMCID: PMC3525664 DOI: 10.1371/journal.pone.0051804] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 11/06/2012] [Indexed: 01/10/2023] Open
Abstract
Trypanosoma cruzi is a protozoan parasite that comprises different phylogenetic groups and is the causative agent of Chagas’ disease. Different T. cruzi strains present differences in infectivity in in vitro and in vivo experimental models, which are likely related to the expression of different virulence factors. Amastin is a surface glycoprotein abundantly expressed on the intracellular mammalian amastigote form of the parasite. In this study, we showed that a highly infective strain (G strain) of extracellular amastigote (EA) invasive forms expressed reduced RNA levels of amastin compared to a less infective strain (CL). The treatment of HeLa cells with recombinant δ-amastin reduced infectivity of EA forms. However, the ectopic expression of δ-amastin accelerated amastigote differentiation into trypomastigotes. Corroborating the virulence behavior in association with amastin expression, the EAs overexpressing amastin were precociously and robustly observed in the liver of susceptible mouse strains (A/JUnib), whereas parasitemia was never detected in in vivo assays. This is the first report on the regulatory role of amastin in the course of both in vitro and in vivo T. cruzi infection.
Collapse
Affiliation(s)
- Mário C. Cruz
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina–UNIFESP, São Paulo, São Paulo, Brazil
| | - Normanda Souza-Melo
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - Claudio Vieira da Silva
- Disciplina de Imunologia - Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Campus Umuarama, Uberlândia, Minas Gerais, Brazil
| | - Wanderson Duarte DaRocha
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - Diana Bahia
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina–UNIFESP, São Paulo, São Paulo, Brazil
| | - Patrícia R. Araújo
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Pampulha, Belo Horizonte, Minas Gerais, Brazil
| | - Santuza R. Teixeira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Pampulha, Belo Horizonte, Minas Gerais, Brazil
| | - Renato A. Mortara
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina–UNIFESP, São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
29
|
Rodrigues AA, Clemente TM, dos Santos MA, Machado FC, Gomes RGB, Moreira HHT, Cruz MC, Brígido PC, dos Santos PCF, Martins FA, Bahia D, Maricato JT, Janini LMR, Reboredo EH, Mortara RA, da Silva CV. A recombinant protein based on Trypanosoma cruzi P21 enhances phagocytosis. PLoS One 2012; 7:e51384. [PMID: 23251513 PMCID: PMC3519637 DOI: 10.1371/journal.pone.0051384] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 11/06/2012] [Indexed: 12/12/2022] Open
Abstract
Background P21 is a secreted protein expressed in all developmental stages of Trypanosoma cruzi. The aim of this study was to determine the effect of the recombinant protein based on P21 (P21-His6) on inflammatory macrophages during phagocytosis. Findings Our results showed that P21-His6 acts as a phagocytosis inducer by binding to CXCR4 chemokine receptor and activating actin polymerization in a way dependent onthe PI3-kinase signaling pathway. Conclusions Thus, our results shed light on the notion that native P21 is a component related to T. cruzi evasion from the immune response and that CXCR4 may be involved in phagocytosis. P21-His6 represents an important experimental control tool to study phagocytosis signaling pathways of different intracellular parasites and particles.
Collapse
Affiliation(s)
- Adele A. Rodrigues
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Tatiana M. Clemente
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Marlus A. dos Santos
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Fabrício C. Machado
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Rafael G. B. Gomes
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | | | - Mário C. Cruz
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina - Universidade Federal de São Paulo, São Paulo, Brazil
| | - Paula C. Brígido
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Paulo C. F. dos Santos
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Flávia A. Martins
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Diana Bahia
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina - Universidade Federal de São Paulo, São Paulo, Brazil
| | - Juliana T. Maricato
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina - Universidade Federal de São Paulo, São Paulo, Brazil
| | - Luiz M. R. Janini
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina - Universidade Federal de São Paulo, São Paulo, Brazil
| | - Eduardo H. Reboredo
- Instituto de Física de São Carlos, Universidade de São Paulo, São Carlos, Brazil
| | - Renato A. Mortara
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina - Universidade Federal de São Paulo, São Paulo, Brazil
| | - Claudio V. da Silva
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
- * E-mail:
| |
Collapse
|
30
|
Ferreira ER, Bonfim-Melo A, Mortara RA, Bahia D. Trypanosoma cruzi extracellular amastigotes and host cell signaling: more pieces to the puzzle. Front Immunol 2012; 3:363. [PMID: 23264776 PMCID: PMC3525110 DOI: 10.3389/fimmu.2012.00363] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 11/12/2012] [Indexed: 11/13/2022] Open
Abstract
Among the different infective stages that Trypanosoma cruzi employs to invade cells, extracellular amastigotes (EAs) have recently gained attention by our group. This is true primarily because these amastigotes are able to infect cultured cells and animals, establishing a sustainable infective cycle. EAs are thus an excellent means of adaptation and survival for T. cruzi, whose different infective stages each utilize unique mechanisms for attachment and penetration. Here we discuss some features of host cell invasion by EAs and the associated host cell signaling events that occur as part of the process.
Collapse
Affiliation(s)
- Eden R Ferreira
- Disciplina de Parasitologia, Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo São Paulo, SP, Brazil
| | | | | | | |
Collapse
|
31
|
IFN-γ plays a unique role in protection against low virulent Trypanosoma cruzi strain. PLoS Negl Trop Dis 2012; 6:e1598. [PMID: 22509418 PMCID: PMC3317909 DOI: 10.1371/journal.pntd.0001598] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2011] [Accepted: 02/23/2012] [Indexed: 12/04/2022] Open
Abstract
Background T. cruzi strains have been divided into six discrete typing units (DTUs) according to their genetic background. These groups are designated T. cruzi I to VI. In this context, amastigotes from G strain (T. cruzi I) are highly infective in vitro and show no parasitemia in vivo. Here we aimed to understand why amastigotes from G strain are highly infective in vitro and do not contribute for a patent in vivo infection. Methodology/Principal Findings Our in vitro studies demonstrated the first evidence that IFN-γ would be associated to the low virulence of G strain in vivo. After intraperitoneal amastigotes inoculation in wild-type and knockout mice for TNF-α, Nod2, Myd88, iNOS, IL-12p40, IL-18, CD4, CD8 and IFN-γ we found that the latter is crucial for controlling infection by G strain amastigotes. Conclusions/Significance Our results showed that amastigotes from G strain are highly infective in vitro but did not contribute for a patent infection in vivo due to its susceptibility to IFN-γ production by host immune cells. These data are useful to understand the mechanisms underlying the contrasting behavior of different T. cruzi groups for in vitro and in vivo infection. Trypanosoma cruzi, an obligate intracellular protozoan, is the etiological agent of Chagas disease that represents an important public health burden in Latin America. The infection with this parasite can lead to severe complications in cardiac and gastrointestinal tissue depending on the strain of parasite and host genetics. Currently, six genetic groups (T. cruzi I to VI) have been identified in this highly genetic and diverse parasite.The majority of published data concerning host immune response has been obtained from studying T. cruzi II to VI-infected mice, and the genetic differences between T. cruzi II to VI and T. cruzi I strains are large. Here we aimed to understand how amastigotes from T. cruzi I G strain are highly infective in vitro and do not contribute for a patent parasitemia in vivo. Our results showed that amastigotes from G strain are highly susceptible to IFN-γ treatment in vitro and secretion by immune cells in vivo. This information may represent important findings to design novel immune strategies to control pathology that may be caused by different strains in the same host.
Collapse
|
32
|
Epigenetic regulation of transcription and virulence in Trypanosoma cruzi by O-linked thymine glucosylation of DNA. Mol Cell Biol 2011; 31:1690-700. [PMID: 21321080 DOI: 10.1128/mcb.01277-10] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Unlike other eukaryotes, the protein-coding genes of Trypanosoma cruzi are arranged in large polycistronic gene clusters transcribed by polymerase II (Pol II). Thus, it is thought that trypanosomes rely solely on posttranscriptional processes to regulate gene expression. Here, we show that the glucosylated thymine DNA base (β-d-glucosyl-hydroxymethyluracil or base J) is present within sequences flanking the polycistronic units (PTUs) in T. cruzi. The loss of base J at sites of transcription initiation, via deletion of the two enzymes that regulate base J synthesis (JBP1 and JBP2), correlates with an increased rate of Pol II transcription and subsequent genome-wide increase in gene expression. The affected genes include virulence genes, and the resulting parasites are defective in host cell invasion and egress. These studies indicate that base J is an epigenetic factor regulating Pol II transcription initiation in kinetoplastids and provides the first biological role of the only hypermodified DNA base in eukaryotes.
Collapse
|
33
|
The challenge of Chagas’ disease: Has the human pathogen, Trypanosoma cruzi, learned how to modulate signaling events to subvert host cells? N Biotechnol 2010; 27:837-43. [DOI: 10.1016/j.nbt.2010.02.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2009] [Accepted: 02/14/2010] [Indexed: 12/20/2022]
|
34
|
Review on Trypanosoma cruzi: Host Cell Interaction. Int J Cell Biol 2010; 2010. [PMID: 20811486 PMCID: PMC2926652 DOI: 10.1155/2010/295394] [Citation(s) in RCA: 161] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Revised: 05/11/2010] [Accepted: 06/04/2010] [Indexed: 12/21/2022] Open
Abstract
Trypanosoma cruzi, the causative agent of Chagas' disease, which affects a large number of individuals in Central and South America, is transmitted to vertebrate hosts by blood-sucking insects. This protozoan is an obligate intracellular parasite. The infective forms of the parasite are metacyclic and bloodstream trypomastigote and amastigote. Metacyclic trypomastigotes are released with the feces of the insect while amastigotes and bloodstream trypomastigotes are released from the infected host cells of the vertebrate host after a complex intracellular life cycle. The recognition between parasite and mammalian host cell involves numerous molecules present in both cell types. Here, we present a brief review of the interaction between Trypanosoma cruzi and its host cells, mainly emphasizing the mechanisms and molecules that participate in the T. cruzi invasion process of the mammalian cells.
Collapse
|
35
|
Alves MJM, Mortara RA. A century of research: what have we learned about the interaction of Trypanosoma cruzi with host cells? Mem Inst Oswaldo Cruz 2009; 104 Suppl 1:76-88. [DOI: 10.1590/s0074-02762009000900013] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Accepted: 05/29/2009] [Indexed: 12/31/2022] Open
|