1
|
Harrison RE, Vogel KJ, Etheridge RD. Stable colonization of the kissing bug Rhodnius prolixus by Trypanosoma cruzi Y strain. PLoS Negl Trop Dis 2025; 19:e0012906. [PMID: 40073048 PMCID: PMC11928063 DOI: 10.1371/journal.pntd.0012906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 03/21/2025] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Trypanosoma cruzi is a single-celled eukaryotic parasite responsible for Chagas disease, a major cause of morbidity and mortality in Central and South America. While the host-pathogen interactions of T. cruzi have been extensively studied in vertebrate models, investigations into its interactions within its insect host remain limited. To address this gap and establish a genetically tractable system for studying parasite-vector dynamics, we conducted quantitative kinetic infection studies using the Y strain of T. cruzi and the model vector Rhodnius prolixus. We began by comparing parasite infection kinetics from two genetically diverse strains of T. cruzi, Brazil and Y, and demonstrated that ingested parasites from both strains transiently expand in the anterior regions of the insect digestive tract with stable colonization occurring in the hindgut over the long term. Notably, we demonstrated that the clonal Y strain, contrary to previous reports, can effectively infect and persist across multiple developmental stages of R. prolixus. Additionally, comparison of movement of parasites versus inert fluorescent microspheres introduced into artificial blood meals suggests that T. cruzi colonization of the R. prolixus gut occurs passively through peristaltic movement during digestion, rather than through active parasite-mediated chemotaxis. These findings highlight the T. cruzi Y strain - R. prolixus model system as a promising tool for the in-depth molecular characterization of parasite-vector interactions, potentially offering new insights into the biology of this neglected and deadly human pathogen.
Collapse
Affiliation(s)
- Ruby E Harrison
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
- Department of Cellular Biology, University of Georgia, Athens, Georgia, United States of America
| | - Kevin J Vogel
- Department of Entomology, University of Georgia, Athens, Georgia, United States of America
| | - Ronald Drew Etheridge
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
- Department of Cellular Biology, University of Georgia, Athens, Georgia, United States of America
| |
Collapse
|
2
|
Jiménez-Ortega RF, Alejandre-Aguilar R, Rivas N, Sánchez F, Sánchez-Muñoz F, Ballinas-Verdugo MA. Ninoa T. cruzi Strain Modifies the Expression of microRNAs in Cardiac Tissue and Plasma During Chagas Disease Infection. Pathogens 2024; 13:1127. [PMID: 39770386 PMCID: PMC11679500 DOI: 10.3390/pathogens13121127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/07/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Chronic chagasic cardiomyopathy is the most severe clinical manifestation of Chagas disease, which affects approximately seven million people worldwide. Latin American countries bear the highest burden, with the greatest morbidity and mortality rates. Currently, diagnostic methods do not provide information on the risk of progression to severe stages of the disease. Recently, microRNAs (miRNAs) have been proposed as promising tools for monitoring the progression of Chagas disease. This study aimed to analyze the expression profiles of the miRNAs miR-1, miR-16, miR-208, and miR-208b in cardiac tissue, plasma, and plasma extracellular vesicles from Ninoa TcI-infected mice during the acute and indeterminate phases of Chagas disease. Methods: The cardiac-specific miRNAs and miR-16 levels were examined in all samples using RT-qPCR. Additionally, pathway analysis was performed to investigate the impact of potential miRNA target genes across various databases. Results: Elevated miR-208b expression was observed in cardiac tissue and plasma during the acute phase. Bioinformatic analysis identified three pathways implicated in disease progression: phosphatidylinositol 3-kinase signaling, Fc gamma receptor-mediated phagocytosis, and leukocyte transendothelial migration, as well as cholinergic synapse pathways. Conclusions: MiR-208b was upregulated during the acute phase and downregulated in the indeterminate phase, suggesting it may play a crucial role in disease progression.
Collapse
Affiliation(s)
- Rogelio F. Jiménez-Ortega
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico;
- Unidad de Acupuntura Rehabilitatoria, Universidad Estatal del Valle de Ecatepec (UNEVE), Ecatepec 55210, Estado de México, Mexico
| | - Ricardo Alejandre-Aguilar
- Departamento de Parasitología, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City 07738, Mexico; (R.A.-A.); (N.R.)
| | - Nancy Rivas
- Departamento de Parasitología, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City 07738, Mexico; (R.A.-A.); (N.R.)
| | - Fausto Sánchez
- División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana Xochimilco (UAM-X), Mexico City 04960, Mexico;
| | - Fausto Sánchez-Muñoz
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez (INCICH), Mexico City 14080, Mexico;
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez (INCICH), Mexico City 14080, Mexico
| | - Martha A. Ballinas-Verdugo
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez (INCICH), Mexico City 14080, Mexico
| |
Collapse
|
3
|
Orlando LMR, Lara LDS, Lechuga GC, Rodrigues GC, Pandoli OG, de Sá DS, Pereira MCDS. Antitrypanosomal Activity of 1,2,3-Triazole-Based Hybrids Evaluated Using In Vitro Preclinical Translational Models. BIOLOGY 2023; 12:1222. [PMID: 37759621 PMCID: PMC10525445 DOI: 10.3390/biology12091222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/08/2023] [Accepted: 07/17/2023] [Indexed: 09/29/2023]
Abstract
Chagas disease therapy still relies on two nitroderivatives, nifurtimox and benznidazole (Bz), which have important limitations and serious adverse effects. New therapeutic alternatives for this silent disease, which has become a worldwide public health problem, are essential for its control and elimination. In this study, 1,2,3-triazole analogues were evaluated for efficacy against T. cruzi. Three triazole derivatives, 1d (0.21 µM), 1f (1.23 µM), and 1g (2.28 µM), showed potent activity against trypomastigotes, reaching IC50 values 10 to 100 times greater than Bz (22.79 µM). Promising candidates are active against intracellular amastigotes (IC50 ≤ 6.20 µM). Treatment of 3D cardiac spheroids, a translational in vitro model, significantly reduced parasite load, indicating good drug diffusion and efficacy. Oral bioavailability was predicted for triazole derivatives. Although infection was significantly reduced without drug pressure in a washout assay, the triazole derivatives did not inhibit parasite resurgence. An isobologram analysis revealed an additive interaction when 1,2,3-triazole analogs and Bz were combined in vitro. These data indicate a strengthened potential of the triazole scaffold and encourage optimization based on an analysis of the structure-activity relationship aimed at identifying new compounds potentially active against T. cruzi.
Collapse
Affiliation(s)
- Lorraine Martins Rocha Orlando
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fiocruz Av. Brasil 4365, Rio de Janeiro 21040-900, Brazil; (L.M.R.O.); (L.d.S.L.); (G.C.L.)
| | - Leonardo da Silva Lara
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fiocruz Av. Brasil 4365, Rio de Janeiro 21040-900, Brazil; (L.M.R.O.); (L.d.S.L.); (G.C.L.)
| | - Guilherme Curty Lechuga
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fiocruz Av. Brasil 4365, Rio de Janeiro 21040-900, Brazil; (L.M.R.O.); (L.d.S.L.); (G.C.L.)
| | - Giseli Capaci Rodrigues
- Programa de Pós-Graduação em Ensino das Ciências, Unigranrio Rua Prof. José de Souza Herdy, Duque de Caxias, Rio de Janeiro 25071-970, Brazil;
| | - Omar Ginoble Pandoli
- Departamento de Química, Pontifícia Universidade Católica, Rua Marquês de São Vincente, 225, Rio de Janeiro 22451-900, Brazil; (O.G.P.); (D.S.d.S.)
- Dipartimento di Farmacia, Università degli Studi di Genova, Viale Cembrano 4, 16126 Genova, Italy
| | - Druval Santos de Sá
- Departamento de Química, Pontifícia Universidade Católica, Rua Marquês de São Vincente, 225, Rio de Janeiro 22451-900, Brazil; (O.G.P.); (D.S.d.S.)
| | - Mirian Claudia de Souza Pereira
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fiocruz Av. Brasil 4365, Rio de Janeiro 21040-900, Brazil; (L.M.R.O.); (L.d.S.L.); (G.C.L.)
| |
Collapse
|
4
|
Finamore-Araujo P, Silva da Fonseca GL, Vieira CS, de Castro DP, Moreira OC. RNA as a feasible marker of Trypanosoma cruzi viability during the parasite interaction with the triatomine vector Rhodnius prolixus (Hemiptera, Triatominae). PLoS Negl Trop Dis 2022; 16:e0010535. [PMID: 35797352 PMCID: PMC9307183 DOI: 10.1371/journal.pntd.0010535] [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: 12/20/2021] [Revised: 07/22/2022] [Accepted: 05/25/2022] [Indexed: 11/18/2022] Open
Abstract
A recurring question concerning Trypanosoma cruzi DNA detection/quantification is related to the fact that DNA amplification, by itself, does not differentiate between viable or dead parasites. On the other hand, RNA can be considered a potential molecular marker of pathogens viability. Herein, we developed a quantitative real-time PCR with reverse Transcription (RT-qPCR) to quantify viable T. cruzi in artificially infected Rhodnius prolixus whilst evaluating differences between DNA and mRNA quantification along the insect midgut during 5, 9, 15 and 29 days after feeding. The RT-qPCR presented an improved performance with linearities ranging from 107 to 102 parasites equivalents and 3 to 0.0032 intestine unit equivalents, and efficiencies of 100.3% and 102.8% for both T. cruzi and triatomine targets, respectively. Comparing both RT-qPCR and qPCR, we confirmed that RNA is faster degraded, no longer being detected at day 1 after parasite lysis, while DNA detection was stable, with no decrease in parasite load over the days, even after parasite lysis. We also observed statistical differences between the quantification of the parasite load by DNA and by RNA on day 15 after feeding of experimentally infected R. prolixus. When assessing different portions of the digestive tract, by RT-qPCR, we could detect a statistically significant reduction in the parasite amount in the anterior midgut. Oppositely, there was a statistically significant increase of the parasite load in the hindgut. In conclusion, for this study parasite’s viability in R. prolixus digestive tract were assessed targeting T. cruzi mRNA. In addition, differences between DNA and RNA detection observed herein, raise the possibility that RNA is a potential molecular viability marker, which could contribute to understanding the dynamics of the parasite infection in invertebrate hosts. In this study, we developed and standardized a Real-Time PCR with Reverse Transcription (RT-qPCR) to determine T. cruzi viability in R. prolixus samples. Moreover, we aimed to assess differences between the amplification signals of DNA and mRNA on a T. cruzi colonization kinetics in experimentally infected R. prolixus. Thus, it was possible to analyze the potential of parasite’s RNA as a molecular viability marker in parasite-vector interaction. This novel RT-qPCR methodology has potential application in viability assessment and raises the possibility for further monitoring of the parasite load in infected insects or studies related to vectorial capacity. Furthermore, the analysis of parasite viability by RT qPCR could be an especially effective tool for Chagas disease diagnostic purposes.
Collapse
Affiliation(s)
- Paula Finamore-Araujo
- Real Time PCR Platform RPT09A, Laboratory of Molecular Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Gabriel Lucio Silva da Fonseca
- Real Time PCR Platform RPT09A, Laboratory of Molecular Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Cecília Stahl Vieira
- Laboratory of Biochemistry and Physiology of Insects, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Postgraduate Program in Science and Biotechnology, Biology Institute, Federal Fluminense University, Niterói, Brazil
| | - Daniele Pereira de Castro
- Laboratory of Biochemistry and Physiology of Insects, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Otacilio Cruz Moreira
- Real Time PCR Platform RPT09A, Laboratory of Molecular Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
5
|
Lara LDS, Lechuga GC, Orlando LMR, Ferreira BS, Souto BA, dos Santos MS, Pereira MCDS. Bioactivity of Novel Pyrazole-Thiazolines Scaffolds against Trypanosoma cruzi: Computational Approaches and 3D Spheroid Model on Drug Discovery for Chagas Disease. Pharmaceutics 2022; 14:995. [PMID: 35631581 PMCID: PMC9146228 DOI: 10.3390/pharmaceutics14050995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 12/01/2022] Open
Abstract
Chagas disease, a century-old disease that mainly affects the impoverished population in Latin America, causes high morbidity and mortality in endemic countries. The available drugs, benznidazole (Bz) and nifurtimox, have limited effectiveness and intense side effects. Drug repurposing, and the development of new chemical entities with potent activity against Trypanosoma cruzi, are a potential source of therapeutic options. The present study describes the biological activity of two new series of pyrazole-thiazoline derivatives, based on optimization of a hit system 5-aminopyrazole-imidazoline previously identified, using structure−activity relationship exploration, and computational and phenotype-based strategies. Promising candidates, 2c, 2e, and 2i derivatives, showed good oral bioavailability and ADMET properties, and low cytotoxicity (CC50 > 100 µM) besides potent activity against trypomastigotes (0.4−2.1 µM) compared to Bz (19.6 ± 2.3 µM). Among them, 2c also stands out, with greater potency against intracellular amastigotes (pIC50 = 5.85). The selected pyrazole-thiazoline derivatives showed good permeability and effectiveness in the 3D spheroids system, but did not sustain parasite clearance in a washout assay. The compounds’ mechanism of action is still unknown, since the treatment neither increased reactive oxygen species, nor reduced cysteine protease activity. This new scaffold will be targeted to optimize in order to enhance its biological activity to identify new drug candidates for Chagas disease therapy.
Collapse
Affiliation(s)
- Leonardo da Silva Lara
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fiocruz, Av. Brasil 4365, Manguinhos, Rio de Janeiro 21040-900, Brazil; (L.d.S.L.); (G.C.L.); (L.M.R.O.)
| | - Guilherme Curty Lechuga
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fiocruz, Av. Brasil 4365, Manguinhos, Rio de Janeiro 21040-900, Brazil; (L.d.S.L.); (G.C.L.); (L.M.R.O.)
| | - Lorraine Martins Rocha Orlando
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fiocruz, Av. Brasil 4365, Manguinhos, Rio de Janeiro 21040-900, Brazil; (L.d.S.L.); (G.C.L.); (L.M.R.O.)
| | - Byanca Silva Ferreira
- Laboratório de Síntese de Sistemas Heterocíclicos (LaSSH), Instituto de Física e Química, Universidade Federal de Itajubá, Avenida BPS, 1303, Pinheirinho, Itajubá 37500-903, Brazil; (B.S.F.); (B.A.S.); (M.S.d.S.)
| | - Bernardo Araújo Souto
- Laboratório de Síntese de Sistemas Heterocíclicos (LaSSH), Instituto de Física e Química, Universidade Federal de Itajubá, Avenida BPS, 1303, Pinheirinho, Itajubá 37500-903, Brazil; (B.S.F.); (B.A.S.); (M.S.d.S.)
| | - Maurício Silva dos Santos
- Laboratório de Síntese de Sistemas Heterocíclicos (LaSSH), Instituto de Física e Química, Universidade Federal de Itajubá, Avenida BPS, 1303, Pinheirinho, Itajubá 37500-903, Brazil; (B.S.F.); (B.A.S.); (M.S.d.S.)
| | - Mirian Claudia de Souza Pereira
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fiocruz, Av. Brasil 4365, Manguinhos, Rio de Janeiro 21040-900, Brazil; (L.d.S.L.); (G.C.L.); (L.M.R.O.)
| |
Collapse
|
6
|
Structural Optimization and Biological Activity of Pyrazole Derivatives: Virtual Computational Analysis, Recovery Assay and 3D Culture Model as Potential Predictive Tools of Effectiveness against Trypanosoma cruzi. Molecules 2021; 26:molecules26216742. [PMID: 34771151 PMCID: PMC8587750 DOI: 10.3390/molecules26216742] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 12/18/2022] Open
Abstract
Chagas disease, a chronic and silent disease caused by Trypanosoma cruzi, is currently a global public health problem. The treatment of this neglected disease relies on benznidazole and nifurtimox, two nitroheterocyclic drugs that show limited efficacy and severe side effects. The failure of potential drug candidates in Chagas disease clinical trials highlighted the urgent need to identify new effective chemical entities and more predictive tools to improve translational success in the drug development pipeline. In this study, we designed a small library of pyrazole derivatives (44 analogs) based on a hit compound, previously identified as a T. cruzi cysteine protease inhibitor. The in vitro phenotypic screening revealed compounds 3g, 3j, and 3m as promising candidates, with IC50 values of 6.09 ± 0.52, 2.75 ± 0.62, and 3.58 ± 0.25 µM, respectively, against intracellular amastigotes. All pyrazole derivatives have good oral bioavailability prediction. The structure–activity relationship (SAR) analysis revealed increased potency of 1-aryl-1H-pyrazole-imidazoline derivatives with the Br, Cl, and methyl substituents in the para-position. The 3m compound stands out for its trypanocidal efficacy in 3D microtissue, which mimics tissue microarchitecture and physiology, and abolishment of parasite recrudescence in vitro. Our findings encourage the progression of the promising candidate for preclinical in vivo studies.
Collapse
|
7
|
Zuma AA, de Souza W. Chagas Disease Chemotherapy: What Do We Know So Far? Curr Pharm Des 2021; 27:3963-3995. [PMID: 33593251 DOI: 10.2174/1381612827666210216152654] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 01/13/2021] [Indexed: 11/22/2022]
Abstract
Chagas disease is a Neglected Tropical Disease (NTD), and although endemic in Latin America, affects around 6-7 million people infected worldwide. The treatment of Chagas disease is based on benznidazole and nifurtimox, which are the only available drugs. However, they are not effective during the chronic phase and cause several side effects. Furthermore, BZ promotes cure in 80% of the patients in the acute phase, but the cure rate drops to 20% in adults in the chronic phase of the disease. In this review, we present several studies published in the last six years, which describes the antiparasitic potential of distinct drugs, from the synthesis of new compounds aiming to target the parasite, as well as the repositioning and the combination of drugs. We highlight several compounds for having shown results that are equivalent or superior to BZ, which means that they should be further studied, either in vitro or in vivo. Furthermore, we stand out the differences in the effects of BZ on the same strain of T. cruzi, which might be related to methodological differences such as parasite and cell ratios, host cell type and the time of adding the drug. In addition, we discuss the wide variety of strains and also the cell types used as a host cell, which makes it difficult to compare the trypanocidal effect of the compounds.
Collapse
Affiliation(s)
- Aline Araujo Zuma
- Laboratorio de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro. Av. Carlos Chagas Filho, 373, Centro de Ciências da Saúde, Cidade Universitária, Ilha do Fundão, 21491-590, Rio de Janeiro, RJ. Brazil
| | - Wanderley de Souza
- Laboratorio de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro. Av. Carlos Chagas Filho, 373, Centro de Ciências da Saúde, Cidade Universitária, Ilha do Fundão, 21491-590, Rio de Janeiro, RJ. Brazil
| |
Collapse
|
8
|
Ventura JD, Beloor J, Allen E, Zhang T, Haugh KA, Uchil PD, Ochsenbauer C, Kieffer C, Kumar P, Hope TJ, Mothes W. Longitudinal bioluminescent imaging of HIV-1 infection during antiretroviral therapy and treatment interruption in humanized mice. PLoS Pathog 2019; 15:e1008161. [PMID: 31805155 PMCID: PMC6917343 DOI: 10.1371/journal.ppat.1008161] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 12/17/2019] [Accepted: 10/29/2019] [Indexed: 12/24/2022] Open
Abstract
Non-invasive bioluminescent imaging (NIBLI) of HIV-1 infection dynamics allows for real-time monitoring of viral spread and the localization of infected cell populations in living animals. In this report, we describe full-length replication-competent GFP and Nanoluciferase (Nluc) expressing HIV-1 reporter viruses from two clinical transmitted / founder (T/F) strains: TRJO.c and Q23.BG505. By infecting humanized mice with these HIV-1 T/F reporter viruses, we were able to directly monitor longitudinal viral spread at whole-animal resolution via NIBLI at a sensitivity of as few as 30-50 infected cells. Bioluminescent signal strongly correlated with HIV-1 infection and responded proportionally to virus suppression in vivo in animals treated daily with a combination antiretroviral therapy (cART) regimen. Longitudinal NIBLI following cART withdrawal visualized tissue-sites that harbored virus during infection recrudescence. Notably, we observed rebounding infection in the same lymphoid tissues where infection was first observed prior to ART treatment. Our work demonstrates the utility of our system for studying in vivo viral infection dynamics and identifying infected tissue regions for subsequent analyses.
Collapse
Affiliation(s)
- John D. Ventura
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, United States of America
| | - Jagadish Beloor
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States of America
| | - Edward Allen
- Department of Cellular and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States of America
| | - Tongyu Zhang
- School of Molecular and Cellular Biology, College of Liberal Arts and Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
| | - Kelsey A. Haugh
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, United States of America
| | - Pradeep D. Uchil
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, United States of America
| | - Christina Ochsenbauer
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Collin Kieffer
- School of Molecular and Cellular Biology, College of Liberal Arts and Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
| | - Priti Kumar
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States of America
| | - Thomas J. Hope
- Department of Cellular and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States of America
| | - Walther Mothes
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, United States of America
| |
Collapse
|
9
|
Avci P, Karimi M, Sadasivam M, Antunes-Melo WC, Carrasco E, Hamblin MR. In-vivo monitoring of infectious diseases in living animals using bioluminescence imaging. Virulence 2017; 9:28-63. [PMID: 28960132 PMCID: PMC6067836 DOI: 10.1080/21505594.2017.1371897] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Traditional methods of localizing and quantifying the presence of pathogenic microorganisms in living experimental animal models of infections have mostly relied on sacrificing the animals, dissociating the tissue and counting the number of colony forming units. However, the discovery of several varieties of the light producing enzyme, luciferase, and the genetic engineering of bacteria, fungi, parasites and mice to make them emit light, either after administration of the luciferase substrate, or in the case of the bacterial lux operon without any exogenous substrate, has provided a new alternative. Dedicated bioluminescence imaging (BLI) cameras can record the light emitted from living animals in real time allowing non-invasive, longitudinal monitoring of the anatomical location and growth of infectious microorganisms as measured by strength of the BLI signal. BLI technology has been used to follow bacterial infections in traumatic skin wounds and burns, osteomyelitis, infections in intestines, Mycobacterial infections, otitis media, lung infections, biofilm and endodontic infections and meningitis. Fungi that have been engineered to be bioluminescent have been used to study infections caused by yeasts (Candida) and by filamentous fungi. Parasitic infections caused by malaria, Leishmania, trypanosomes and toxoplasma have all been monitored by BLI. Viruses such as vaccinia, herpes simplex, hepatitis B and C and influenza, have been studied using BLI. This rapidly growing technology is expected to continue to provide much useful information, while drastically reducing the numbers of animals needed in experimental studies.
Collapse
Affiliation(s)
- Pinar Avci
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,b Department of Dermatology , Harvard Medical School , Boston , MA , USA
| | - Mahdi Karimi
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,c Department of Medical Nanotechnology , School of Advanced Technologies in Medicine, Iran University of Medical Sciences , Tehran , Iran.,d Cellular and Molecular Research Center, Iran University of Medical Sciences , Tehran , Iran
| | - Magesh Sadasivam
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,e Amity Institute of Nanotechnology, Amity University Uttar Pradesh , Noida , India
| | - Wanessa C Antunes-Melo
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,f University of Sao Paulo , Sao Carlos-SP , Brazil
| | - Elisa Carrasco
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,g Department of Biosciences , Durham University , Durham , United Kingdom
| | - Michael R Hamblin
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,b Department of Dermatology , Harvard Medical School , Boston , MA , USA.,h Harvard-MIT Division of Health Sciences and Technology , Cambridge , MA , USA
| |
Collapse
|
10
|
Silva-dos-Santos D, Barreto-de-Albuquerque J, Guerra B, Moreira OC, Berbert LR, Ramos MT, Mascarenhas BAS, Britto C, Morrot A, Serra Villa-Verde DM, Garzoni LR, Savino W, Cotta-de-Almeida V, de Meis J. Unraveling Chagas disease transmission through the oral route: Gateways to Trypanosoma cruzi infection and target tissues. PLoS Negl Trop Dis 2017; 11:e0005507. [PMID: 28379959 PMCID: PMC5397068 DOI: 10.1371/journal.pntd.0005507] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 04/19/2017] [Accepted: 03/20/2017] [Indexed: 02/02/2023] Open
Abstract
Oral transmission of Trypanosoma cruzi, the causative agent of Chagas disease, is the most important route of infection in Brazilian Amazon and Venezuela. Other South American countries have also reported outbreaks associated with food consumption. A recent study showed the importance of parasite contact with oral cavity to induce a highly severe acute disease in mice. However, it remains uncertain the primary site of parasite entry and multiplication due to an oral infection. Here, we evaluated the presence of T. cruzi Dm28c luciferase (Dm28c-luc) parasites in orally infected mice, by bioluminescence and quantitative real-time PCR. In vivo bioluminescent images indicated the nasomaxillary region as the site of parasite invasion in the host, becoming consistently infected throughout the acute phase. At later moments, 7 and 21 days post-infection (dpi), luminescent signal is denser in the thorax, abdomen and genital region, because of parasite dissemination in different tissues. Ex vivo analysis demonstrated that the nasomaxillary region, heart, mandibular lymph nodes, liver, spleen, brain, epididymal fat associated to male sex organs, salivary glands, cheek muscle, mesenteric fat and lymph nodes, stomach, esophagus, small and large intestine are target tissues at latter moments of infection. In the same line, amastigote nests of Dm28c GFP T. cruzi were detected in the nasal cavity of 6 dpi mice. Parasite quantification by real-time qPCR at 7 and 21 dpi showed predominant T. cruzi detection and expansion in mouse nasal cavity. Moreover, T. cruzi DNA was also observed in the mandibular lymph nodes, pituitary gland, heart, liver, small intestine and spleen at 7 dpi, and further, disseminated to other tissues, such as the brain, stomach, esophagus and large intestine at 21 dpi. Our results clearly demonstrated that oral cavity and adjacent compartments is the main target region in oral T. cruzi infection leading to parasite multiplication at the nasal cavity. Oral transmission of Trypanosoma cruzi associated with food/beverage consumption is presently an important route of infection in Brazil and Venezuela. Colombia, Bolivia, Argentina and Ecuador have also reported to have acute cases of Chagas disease transmission through the oral route. Significant studies about this form of T. cruzi infection are largely lacking. In addition to the classic cardiac involvement, orally-infected patient progress to a highly symptomatic disease and increased mortality rate (8–35%), surpassing the calculated mortality produced by the disease resulting from the biting of infected insect vectors (5–10%). Here, we explored by in vivo bioluminescent images, qPCR and fluorescence microscopy the primary site of parasite entry and multiplication in oral infection (OI). Our results clearly demonstrated that the oral cavity is the main T. cruzi target region in OI, leading to parasite multiplication at the nasal cavity and parasite dissemination to the brain and peripheral tissues. Interestingly, facial edema, paraesthesia of the tongue, gingivitis and dry cough were already described in affected patients. These findings might be associated to our present data, which describe for the first time the nasomaxillary region as the main target tissue following oral T. cruzi infection.
Collapse
Affiliation(s)
- Danielle Silva-dos-Santos
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | | - Bárbara Guerra
- National Center of Structural Biology and Bio-imaging—CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Otacilio C. Moreira
- Laboratory on Molecular Biology and Endemic Diseases, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Luiz Ricardo Berbert
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Mariana Tavares Ramos
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | | - Constança Britto
- Laboratory on Molecular Biology and Endemic Diseases, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Alexandre Morrot
- Department of Immunology, Microbiology Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Déa M. Serra Villa-Verde
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Luciana Ribeiro Garzoni
- Laboratory for Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Wilson Savino
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Vinícius Cotta-de-Almeida
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Juliana de Meis
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
11
|
Azambuja P, Garcia ES, Waniek PJ, Vieira CS, Figueiredo MB, Gonzalez MS, Mello CB, Castro DP, Ratcliffe NA. Rhodnius prolixus: from physiology by Wigglesworth to recent studies of immune system modulation by Trypanosoma cruzi and Trypanosoma rangeli. JOURNAL OF INSECT PHYSIOLOGY 2017; 97:45-65. [PMID: 27866813 DOI: 10.1016/j.jinsphys.2016.11.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 11/04/2016] [Accepted: 11/16/2016] [Indexed: 06/06/2023]
Abstract
This review is dedicated to the memory of Professor Sir Vincent B. Wigglesworth (VW) in recognition of his many pioneering contributions to insect physiology which, even today, form the basis of modern-day research in this field. Insects not only make vital contributions to our everyday lives by their roles in pollination, balancing eco-systems and provision of honey and silk products, but they are also outstanding models for studying the pathogenicity of microorganisms and the functioning of innate immunity in humans. In this overview, the immune system of the triatomine bug, Rhodnius prolixus, is considered which is most appropriate to this dedication as this insect species was the favourite subject of VW's research. Herein are described recent developments in knowledge of the functioning of the R. prolixus immune system. Thus, the roles of the cellular defences, such as phagocytosis and nodule formation, as well as the role of eicosanoids, ecdysone, antimicrobial peptides, reactive oxygen and nitrogen radicals, and the gut microbiota in the immune response of R. prolixus are described. The details of many of these were unknown to VW although his work gives indications of his awareness of the importance to R. prolixus of cellular immunity, antibacterial activity, prophenoloxidase and the gut microbiota. This description of R. prolixus immunity forms a backdrop to studies on the interaction of the parasitic flagellates, Trypanosoma cruzi and Trypanosoma rangeli, with the host defences of this important insect vector. These parasites remarkably utilize different strategies to avoid/modulate the triatomine immune response in order to survive in the extremely hostile host environments present in the vector gut and haemocoel. Much recent information has also been gleaned on the remarkable diversity of the immune system in the R. prolixus gut and its interaction with trypanosome parasites. This new data is reviewed and gaps in our knowledge of R. prolixus immunity are identified as subjects for future endeavours. Finally, the publication of the T. cruzi, T. rangeli and R. prolixus genomes, together with the use of modern molecular techniques, should lead to the enhanced identification of the determinants of infection derived from both the vector and the parasites which, in turn, could form targets for new molecular-based control strategies.
Collapse
Affiliation(s)
- P Azambuja
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, RJ, Brazil; Departamento de Entomologia Molecular, Instituto Nacional de Entomologia Molecular (INCT-EM), Rio de Janeiro, RJ, Brazil.
| | - E S Garcia
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, RJ, Brazil; Departamento de Entomologia Molecular, Instituto Nacional de Entomologia Molecular (INCT-EM), Rio de Janeiro, RJ, Brazil.
| | - P J Waniek
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, RJ, Brazil.
| | - C S Vieira
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, RJ, Brazil.
| | - M B Figueiredo
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, RJ, Brazil.
| | - M S Gonzalez
- Laboratório de Biologia de Insetos, Universidade Federal Fluminense, Niterói, RJ, Brazil.
| | - C B Mello
- Laboratório de Biologia de Insetos, Universidade Federal Fluminense, Niterói, RJ, Brazil.
| | - D P Castro
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, RJ, Brazil; Departamento de Entomologia Molecular, Instituto Nacional de Entomologia Molecular (INCT-EM), Rio de Janeiro, RJ, Brazil.
| | - N A Ratcliffe
- Laboratório de Biologia de Insetos, Universidade Federal Fluminense, Niterói, RJ, Brazil; Department of Biosciences, College of Science, Swansea University, Singleton Park, Swansea, Wales, United Kingdom.
| |
Collapse
|
12
|
Neumann ADS, Dias FDA, Ferreira JDS, Fontes ANB, Rosa PS, Macedo RE, Oliveira JH, Teixeira RLDF, Pessolani MCV, Moraes MO, Suffys PN, Oliveira PL, Sorgine MHF, Lara FA. Experimental Infection of Rhodnius prolixus (Hemiptera, Triatominae) with Mycobacterium leprae Indicates Potential for Leprosy Transmission. PLoS One 2016; 11:e0156037. [PMID: 27203082 PMCID: PMC4874629 DOI: 10.1371/journal.pone.0156037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 05/09/2016] [Indexed: 11/16/2022] Open
Abstract
Leprosy is a chronic dermato-neurological disease caused by infection with Mycobacterium leprae. In 2013 almost 200,000 new cases of leprosy were detected around the world. Since the first symptoms take from years to decades to appear, the total number of asymptomatic patients is impossible to predict. Although leprosy is one of the oldest records of human disease, the mechanisms involved with its transmission and epidemiology are still not completely understood. In the present work, we experimentally investigated the hypothesis that the mosquitoes Aedes aegypti and Culex quinquefasciatus and the hemiptera Rhodnius prolixus act as leprosy vectors. By means of real-time PCR quantification of M. leprae 16SrRNA, we found that M. leprae remained viable inside the digestive tract of Rhodnius prolixus for 20 days after oral infection. In contrast, in the gut of both mosquito species tested, we were not able to detect M. leprae RNA after a similar period of time. Inside the kissing bug Rhodnius prolixus digestive tract, M. leprae was initially restricted to the anterior midgut, but gradually moved towards the hindgut, in a time course reminiscent of the life cycle of Trypanosoma cruzi, a well-known pathogen transmitted by this insect. The maintenance of M. leprae infectivity inside the digestive tract of this kissing bug is further supported by successful mice footpad inoculation with feces collected 20 days after infection. We conclude that Rhodnius prolixus defecate infective M. leprae, justifying the evaluation of the presence of M. leprae among sylvatic and domestic kissing bugs in countries endemic for leprosy.
Collapse
Affiliation(s)
- Arthur da Silva Neumann
- Laboratório de Microbiologia Celular, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Felipe de Almeida Dias
- Laboratório de Bioquímica de Artrópodes Hematófagos, Federal University of Rio de Janeiro, Rio de Janiero, Brazil
| | - Jéssica da Silva Ferreira
- Laboratório de Microbiologia Celular, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Amanda Nogueira Brum Fontes
- Laboratório de Biologia Molecular Aplicada a Micobactérias, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | | - Rafael Enrique Macedo
- Laboratório de Microbiologia Celular, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - José Henrique Oliveira
- Laboratório de Bioquímica de Artrópodes Hematófagos, Federal University of Rio de Janeiro, Rio de Janiero, Brazil
| | | | | | - Milton Ozório Moraes
- Laboratório de Hanseníase, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Philip Noel Suffys
- Laboratório de Biologia Molecular Aplicada a Micobactérias, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Pedro L. Oliveira
- Laboratório de Bioquímica de Artrópodes Hematófagos, Federal University of Rio de Janeiro, Rio de Janiero, Brazil
| | | | - Flavio Alves Lara
- Laboratório de Microbiologia Celular, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
13
|
Trypanosoma cruzi-Trypanosoma rangeli co-infection ameliorates negative effects of single trypanosome infections in experimentally infected Rhodnius prolixus. Parasitology 2016; 143:1157-67. [PMID: 27174360 DOI: 10.1017/s0031182016000615] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Trypanosoma cruzi, causative agent of Chagas disease, co-infects its triatomine vector with its sister species Trypanosoma rangeli, which shares 60% of its antigens with T. cruzi. Additionally, T. rangeli has been observed to be pathogenic in some of its vector species. Although T. cruzi-T. rangeli co-infections are common, their effect on the vector has rarely been investigated. Therefore, we measured the fitness (survival and reproduction) of triatomine species Rhodnius prolixus infected with just T. cruzi, just T. rangeli, or both T. cruzi and T. rangeli. We found that survival (as estimated by survival probability and hazard ratios) was significantly different between treatments, with the T. cruzi treatment group having lower survival than the co-infected treatment. Reproduction and total fitness estimates in the T. cruzi and T. rangeli treatments were significantly lower than in the co-infected and control groups. The T. cruzi and T. rangeli treatment group fitness estimates were not significantly different from each other. Additionally, co-infected insects appeared to tolerate higher doses of parasites than insects with single-species infections. Our results suggest that T. cruzi-T. rangeli co-infection could ameliorate negative effects of single infections of either parasite on R. prolixus and potentially help it to tolerate higher parasite doses.
Collapse
|
14
|
Ferreira BL, Orikaza CM, Cordero EM, Mortara RA. Trypanosoma cruzi: single cell live imaging inside infected tissues. Cell Microbiol 2016; 18:779-83. [PMID: 26639617 PMCID: PMC5064609 DOI: 10.1111/cmi.12553] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 11/03/2015] [Accepted: 12/03/2015] [Indexed: 01/30/2023]
Abstract
Although imaging the live Trypanosoma cruzi parasite is a routine technique in most laboratories, identification of the parasite in infected tissues and organs has been hindered by their intrinsic opaque nature. We describe a simple method for in vivo observation of live single-cell Trypanosoma cruzi parasites inside mammalian host tissues. BALB/c or C57BL/6 mice infected with DsRed-CL or GFP-G trypomastigotes had their organs removed and sectioned with surgical blades. Ex vivo organ sections were observed under confocal microscopy. For the first time, this procedure enabled imaging of individual amastigotes, intermediate forms and motile trypomastigotes within infected tissues of mammalian hosts.
Collapse
Affiliation(s)
- Bianca Lima Ferreira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM-UNIFESP), Rua Botucatu 862 6th floor, São Paulo, São Paulo, Brazil
| | - Cristina Mary Orikaza
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM-UNIFESP), Rua Botucatu 862 6th floor, São Paulo, São Paulo, Brazil
| | - Esteban Mauricio Cordero
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM-UNIFESP), Rua Botucatu 862 6th floor, São Paulo, São Paulo, Brazil
| | - Renato Arruda Mortara
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM-UNIFESP), Rua Botucatu 862 6th floor, São Paulo, São Paulo, Brazil
| |
Collapse
|
15
|
Dias FDA, Guerra B, Vieira LR, Perdomo HD, Gandara ACP, do Amaral RJV, Vollú RE, Gomes SAO, Lara FA, Sorgine MHF, Medei E, de Oliveira PL, Salmon D. Monitoring of the Parasite Load in the Digestive Tract of Rhodnius prolixus by Combined qPCR Analysis and Imaging Techniques Provides New Insights into the Trypanosome Life Cycle. PLoS Negl Trop Dis 2015; 9:e0004186. [PMID: 26496442 PMCID: PMC4619730 DOI: 10.1371/journal.pntd.0004186] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 10/01/2015] [Indexed: 01/12/2023] Open
Abstract
Background Here we report the monitoring of the digestive tract colonization of Rhodnius prolixus by Trypanosoma cruzi using an accurate determination of the parasite load by qPCR coupled with fluorescence and bioluminescence imaging (BLI). These complementary methods revealed critical steps necessary for the parasite population to colonize the insect gut and establish vector infection. Methodology/Principal Findings qPCR analysis of the parasite load in the insect gut showed several limitations due mainly to the presence of digestive-derived products that are thought to degrade DNA and inhibit further the PCR reaction. We developed a real-time PCR strategy targeting the T. cruzi repetitive satellite DNA sequence using as internal standard for normalization, an exogenous heterologous DNA spiked into insect samples extract, to precisely quantify the parasite load in each segment of the insect gut (anterior midgut, AM, posterior midgut, PM, and hindgut, H). Using combined fluorescence microscopy and BLI imaging as well as qPCR analysis, we showed that during their journey through the insect digestive tract, most of the parasites are lysed in the AM during the first 24 hours independently of the gut microbiota. During this short period, live parasites move through the PM to establish the onset of infection. At days 3–4 post-infection (p.i.), the parasite population begins to colonize the H to reach a climax at day 7 p.i., which is maintained during the next two weeks. Remarkably, the fluctuation of the parasite number in H remains relatively stable over the two weeks after refeeding, while the populations residing in the AM and PM increases slightly and probably constitutes the reservoirs of dividing epimastigotes. Conclusions/Significance These data show that a tuned dynamic control of the population operates in the insect gut to maintain an equilibrium between non-dividing infective trypomastigote forms and dividing epimastigote forms of the parasite, which is crucial for vector competence. Although the key aspects of the T. cruzi life cycle were described more than one century ago, the development and interactions of T. cruzi with its vector are poorly characterized. By dissection of different compartments of the triatomine gut (prototype Rhodnius prolixus) (i.e., AM, PM and H) at regular time intervals, we evaluated trypanosome development within the insect using an accurate qPCR assay. qPCR analysis of trypanosomal colonization and clearance dynamics in real-time were confirmed in vivo using both fluorescence and bioluminescence imaging, which revealed massive parasite lysis during the first 24 hours post-feeding (p.f.). After one week, the parasite succeeded in establishing a resident population in each compartment of the gut, albeit at varying levels. From one week after the onset of infection in the AM and PM, some resident forms agglomerated into rosettes, clustering in close association with the vector tissue and constituting potential parasite reservoirs of the bug. For the first time, we have described a methodology to accurately quantify parasites in the insect gut that would be a useful tool for evaluating the impact of RNAi silencing of insect genes during the course of infection by T. cruzi.
Collapse
Affiliation(s)
- Felipe de Almeida Dias
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Barbara Guerra
- Centro Nacional de Biologia Estrutural e Bioimagem—CENABIO, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Larissa Rezende Vieira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Hugo Diego Perdomo
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Caroline Paiva Gandara
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Renata Estebanez Vollú
- Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Flavio Alves Lara
- Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Marcos Henrique Ferreira Sorgine
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Emiliano Medei
- Centro Nacional de Biologia Estrutural e Bioimagem—CENABIO, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro Lagerblad de Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Didier Salmon
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
16
|
Henriques C, Miller MP, Catanho M, de Carvalho TMU, Krieger MA, Probst CM, de Souza W, Degrave W, Amara SG. Identification and functional characterization of a novel arginine/ornithine transporter, a member of a cationic amino acid transporter subfamily in the Trypanosoma cruzi genome. Parasit Vectors 2015; 8:346. [PMID: 26109388 PMCID: PMC4486710 DOI: 10.1186/s13071-015-0950-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 06/13/2015] [Indexed: 01/03/2023] Open
Abstract
Background Trypanosoma cruzi, the etiological agent of Chagas disease, is auxotrophic for arginine. It obtains this amino acid from the host through transporters expressed on the plasma membrane and on the membranes of intracellular compartments. A few cationic amino acid transporters have been characterized at the molecular level, such as the novel intracellular arginine/ornithine transporter, TcCAT1.1, a member of the TcCAT subfamily that is composed of four almost identical open reading frames in the T. cruzi genome. Methods The functional characterization of the TcCAT1.1 isoform was performed in two heterologous expression systems. TcCAT subfamily expression was evaluated by real-time PCR in polysomal RNA fractions, and the cellular localization of TcCAT1.1 fused to EGFP was performed by confocal and immunoelectron microscopy. Results In the S. cerevisiae expression system, TcCAT1.1 showed high affinity for arginine (Km = 0.085 ± 0.04 mM) and low affinity for ornithine (Km = 1.7 ± 0.2 mM). Xenopus laevis oocytes expressing TcCAT1.1 showed a 7-fold increase in arginine uptake when they were pre-loaded with arginine, indicating that transport is enhanced by substrates on the trans side of the membrane (trans-stimulation). Oocytes that were pre-loaded with [3H]-arginine displayed a 16-fold higher efflux of [3H]-arginine compared with that of the control. Analysis of polysomal RNA fractions demonstrated that the expression of members of the arginine transporter TcCAT subfamily is upregulated under nutritional stress and that this upregulation precedes metacyclogenesis. To investigate the cellular localization of the transporter, EGFP was fused to TcCAT1.1, and fluorescence microscopy and immunocytochemistry revealed the intracellular labeling of vesicles in the anterior region, in a network of tubules and vesicles. Conclusions TcCAT1.1 is a novel arginine/ornithine transporter, an exchanger expressed in intracellular compartments that is physiologically involved in arginine homeostasis throughout the T. cruzi life cycle. The properties and estimated kinetic parameters of TcCAT1.1 can be extended to other members of the TcCAT subfamily.
Collapse
Affiliation(s)
- Cristina Henriques
- Fundação Oswaldo Cruz, Fiocruz-Mato Grosso do Sul, Rua Gabriel Abrão 92-Jardim das Nações, Campo Grande, MS, 89081-746, Brazil. .,Instituto de Biofísica Carlos Chagas Filho-UFRJ, CCS-Bloco G-Laboratório de Ultraestrutura Celular Hertha Meyer, Rio de Janeiro, RJ, 21949-900, Brazil. .,Nucleo de Biologia Estrutural e Biomagens, Universidade Federal do Rio de Janeiro-CENABIO, Rio de Janeiro, RJ, Brazil.
| | - Megan P Miller
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15260, USA.
| | - Marcos Catanho
- Fiocruz, Instituto Oswaldo Cruz, Laboratório de Genômica Funcional e Bioinformática, Av. Brasil 4365, Manguinhos, 21040-900, Rio de Janeiro, RJ, Brazil.
| | - Técia Maria Ulisses de Carvalho
- Instituto de Biofísica Carlos Chagas Filho-UFRJ, CCS-Bloco G-Laboratório de Ultraestrutura Celular Hertha Meyer, Rio de Janeiro, RJ, 21949-900, Brazil.
| | | | | | - Wanderley de Souza
- Instituto de Biofísica Carlos Chagas Filho-UFRJ, CCS-Bloco G-Laboratório de Ultraestrutura Celular Hertha Meyer, Rio de Janeiro, RJ, 21949-900, Brazil. .,Nucleo de Biologia Estrutural e Biomagens, Universidade Federal do Rio de Janeiro-CENABIO, Rio de Janeiro, RJ, Brazil. .,Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Biomagens-INBEB, Rio de Janeiro, Brazil.
| | - Wim Degrave
- Fiocruz, Instituto Oswaldo Cruz, Laboratório de Genômica Funcional e Bioinformática, Av. Brasil 4365, Manguinhos, 21040-900, Rio de Janeiro, RJ, Brazil.
| | - Susan Gaye Amara
- National Institute of Mental Health, NIH Building 10 Center Driver, Room 4N222, MSC 1381, Bethesda, MD, 20892-1381, USA. .,Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
17
|
Peterson JK, Graham AL, Dobson AP, Chávez OT. Rhodnius prolixus Life History Outcomes Differ when Infected with Different Trypanosoma cruzi I Strains. Am J Trop Med Hyg 2015; 93:564-72. [PMID: 26078316 DOI: 10.4269/ajtmh.15-0218] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/02/2015] [Indexed: 12/20/2022] Open
Abstract
The effect of a parasite on the life history of its vector is important for understanding and predicting disease transmission. Chagas disease agent Trypanosoma cruzi is a generalist parasite that is diverse across scales from its genetic diversity to the 100s of mammal and vector species it infects. Its vertebrate hosts show quite variable responses to infection, however, to date there are no studies looking at how T. cruzi variability might result in variable outcomes in its invertebrate host. Therefore, we investigated the effect of different T. cruzi I strains on Rhodnius prolixus survival and development. We found significant variation between insects infected with different strains, with some strains having no effect, as compared with uninfected insects, and others with significantly lower survival and development. We also found that different variables had varying importance between strains, with the effect of time postinfection and the blood:weight ratio of the infective meal significantly affecting the survival of insects infected with some strains, but not others. Our results suggest that T. cruzi can be pathogenic not only to its vertebrate hosts but also to its invertebrate hosts.
Collapse
Affiliation(s)
- Jennifer K Peterson
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey; Grupo BCEI, Universidad de Antioquia, Medellín, Colombia
| | - Andrea L Graham
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey; Grupo BCEI, Universidad de Antioquia, Medellín, Colombia
| | - Andrew P Dobson
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey; Grupo BCEI, Universidad de Antioquia, Medellín, Colombia
| | - Omar Triana Chávez
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey; Grupo BCEI, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
18
|
Lewis MD, Francisco AF, Taylor MC, Kelly JM. A new experimental model for assessing drug efficacy against Trypanosoma cruzi infection based on highly sensitive in vivo imaging. ACTA ACUST UNITED AC 2014; 20:36-43. [PMID: 25296657 PMCID: PMC4361455 DOI: 10.1177/1087057114552623] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The protozoan Trypanosoma cruzi is the causative agent of Chagas disease, one of the world’s major neglected infections. Although development of improved antiparasitic drugs is considered a priority, there have been no significant treatment advances in the past 40 years. Factors that have limited progress include an incomplete understanding of pathogenesis, tissue tropism, and disease progression. In addition, in vivo models, which allow parasite burdens to be tracked throughout the chronic stage of infection, have been lacking. To address these issues, we have developed a highly sensitive in vivo imaging system based on bioluminescent T. cruzi, which express a red-shifted luciferase that emits light in the tissue-penetrating orange-red region of the spectrum. The exquisite sensitivity of this noninvasive murine model has been exploited to monitor parasite burden in real time throughout the chronic stage, has allowed the identification of the gastrointestinal tract as the major niche of long-term infection, and has demonstrated that chagasic heart disease can develop in the absence of locally persistent parasites. Here, we review the parameters of the imaging system and describe how this experimental model can be incorporated into drug development programs as a valuable tool for assessing efficacy against both acute and chronic T. cruzi infections.
Collapse
Affiliation(s)
- Michael D Lewis
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Amanda Fortes Francisco
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Martin C Taylor
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - John M Kelly
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| |
Collapse
|
19
|
Henriques C, Henriques-Pons A, Meuser-Batista M, Ribeiro AS, de Souza W. In vivo imaging of mice infected with bioluminescent Trypanosoma cruzi unveils novel sites of infection. Parasit Vectors 2014; 7:89. [PMID: 24589192 PMCID: PMC3973021 DOI: 10.1186/1756-3305-7-89] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 02/21/2014] [Indexed: 11/10/2022] Open
Abstract
Background The development of techniques that allow the imaging of animals infected with parasites expressing luciferase opens up new possibilities for following the fate of parasites in infected mammals. Methods D-luciferin potassium salt stock solution was prepared in phosphate-buffered saline (PBS) at 15 mg/ml. To produce bioluminescence, infected and control mice received an intraperitoneal injection of luciferin stock solution (150 mg/kg). All mice were immediately anesthetized with 2% isofluorane, and after 10 minutes were imaged. Ex vivo evaluation of infected tissues and organs was evaluated in a 24-well plate in 150 μg/ml D-luciferin diluted in PBS. Images were captured using the IVIS Lumina image system (Xenogen). Dissected organs were also evaluated by microscopy of hematoxylin-eosin stained sections. Results Here we describe the results obtained using a genetically modified Dm28c strain of T. cruzi expressing the firefly luciferase to keep track of infection by bioluminescence imaging. Progression of infection was observed in vivo in BALB/c mice at various intervals after infection with transgenic Dm28c-luc. The bioluminescent signal was immediately observed at the site of T. cruzi inoculation, and one day post infection (dpi) it was disseminated in the peritoneal cavity. A similar pattern in the cavity was observed on 7 dpi, but the bioluminescence was more intense in the terminal region of the large intestine, rectum, and gonads. On 14 and 21 dpi, bioluminescent parasites were also observed in the heart, snout, paws, hind limbs, and forelimbs. From 28 dpi to 180 dpi in chronically infected mice, bioluminescence declined in regions of the body but was concentrated in the gonad region. Ex vivo evaluation of dissected organs and tissues by bioluminescent imaging confirmed the in vivo bioluminescent foci. Histopathological analysis of dissected organs demonstrated parasite nests at the rectum and snout, in muscle fibers of mice infected with Dm28c-WT and with Dm28c-luc, corroborating the bioluminescent imaging. Conclusion Bioluminescence imaging is accurate for tracking parasites in vivo, and this methodology is important to gain a better understanding of the infection, tissue inflammation, and parasite biology regarding host cell interaction, proliferation, and parasite clearance to subpatent levels.
Collapse
Affiliation(s)
| | | | | | | | - Wanderley de Souza
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, CCS-Bloco G, Ilha do Fundão, 21941-900 Rio de Janeiro-RJ, Brazil.
| |
Collapse
|
20
|
Goyard S, Dutra PL, Deolindo P, Autheman D, D'Archivio S, Minoprio P. In vivo imaging of trypanosomes for a better assessment of host-parasite relationships and drug efficacy. Parasitol Int 2013; 63:260-8. [PMID: 23892180 DOI: 10.1016/j.parint.2013.07.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 05/20/2013] [Accepted: 07/16/2013] [Indexed: 11/19/2022]
Abstract
The advances in microscopy combined to the invaluable progress carried by the utilization of molecular, immunological or immunochemical markers and the implementation of more powerful imaging technologies have yielded great improvements to the knowledge of the interaction between microorganisms and their hosts, notably a better understanding of the establishment of infectious processes. Still today, the intricacies of the dialog between parasites, cells and tissues remain limited. Some improvements have been attained with the stable integration and expression of the green fluorescence protein or firefly luciferase and other reporter genes, which have allowed to better approach the monitoring of gene expression and protein localization in vivo, in situ and in real time. Aiming at better exploring the well-established models of murine infections with the characterized strains of Trypanosoma cruzi and Trypanosoma vivax, we revisited in the present report the state of the art about the tools for the imaging of Trypanosomatids in vitro and in vivo and show the latest transgenic parasites that we have engineered in our laboratory using conventional transfection methods. The targeting of trypanosomes presented in this study is a promising tool for approaching the biology of parasite interactions with host cells, the progression of the diseases they trigger and the screening of new drugs in vivo or in vitro.
Collapse
Affiliation(s)
- S Goyard
- Institut Pasteur, Laboratoire des Processus Infectieux à Trypanosomatidés, Département Infection et Epidemiologie, 25 rue du Dr. Roux, 75724 Paris, France
| | | | | | | | | | | |
Collapse
|