1
|
Abras A. Isothermal Nucleic Acid Amplification as a Promising and Versatile Diagnostic Approach for Point-of-Care Testing of Congenital Chagas Disease. J Mol Diagn 2024; 26:1042-1044. [PMID: 39332628 DOI: 10.1016/j.jmoldx.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/03/2024] [Accepted: 09/09/2024] [Indexed: 09/29/2024] Open
Affiliation(s)
- Alba Abras
- Àrea de Genètica, Departament de Biologia, Universitat de Girona, Girona, Spain.
| |
Collapse
|
2
|
López Cancino SA, Eligio García L, Crisóstomo Vázquez MDP, Soria Guerrero M, Jiménez Cardoso E, Meneses Mayo M, Islas Andrade SA. Prevalence of Trypanosoma cruzi Infection in Pregnant Women and Risk of Vertical Transmission in Newborns in Chiapas, Mexico. Trop Med Infect Dis 2024; 9:261. [PMID: 39591267 PMCID: PMC11598654 DOI: 10.3390/tropicalmed9110261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 10/26/2024] [Accepted: 10/27/2024] [Indexed: 11/28/2024] Open
Abstract
The Mexican state of Chiapas is considered epidemiologically significant for Chagas disease due to the coexistence of infected reservoirs and vectors, including migratory populations from Central and South America. However, there is a lack of monitoring programs for the timely detection of this disease. The objective of this study was to elucidate the prevalence of Trypanosoma cruzi infection in pregnant women and the risk of vertical transmission in newborns at two hospitals located in the Metropolitan Region of Tuxtla Gutierrez, the capital of Chiapas State Mexico. A cross-sectional study was carried out with 193 pregnant women with gestational ages between 32 and 40 weeks, who underwent immunological testing to diagnose Chagas disease. Conventional PCR testing on cord blood revealed the presence of T. cruzi in newborns. The prevalence of T. cruzi infection in pregnant women was 32.12% (95% confidence interval (CI): 0.25, 0.38). The 62 pregnant women who tested positive for Chagas disease gave birth to 63 children, and in 5 newborns (8% (5/62), 95% confidence interval (CI): 0.02, 0.19), PCR tests on umbilical cord blood were positive for T. cruzi. In conclusion, the dataset showed a high prevalence of Chagas disease in the sample of pregnant women studied and a maternal-fetal transmission rate of 8%.
Collapse
Affiliation(s)
- Sury Antonio López Cancino
- Head of Health Research Office, IMSS BIENESTAR State Coordination Chiapas, Health Services of the Mexican Institute of Social Security for Welfare (IMSS-BIENESTAR), 16 South West 333, Tuxtla Gutiérrez 29067, Mexico
| | - Leticia Eligio García
- Parasitology Research Laboratory, Children Hospital of México “Federico Gómez”, Dr. Márquez 162, Col Doctores, Cuauhtémoc, Mexico City 06720, Mexico; (L.E.G.); (M.d.P.C.V.); (M.S.G.)
| | - María del Pilar Crisóstomo Vázquez
- Parasitology Research Laboratory, Children Hospital of México “Federico Gómez”, Dr. Márquez 162, Col Doctores, Cuauhtémoc, Mexico City 06720, Mexico; (L.E.G.); (M.d.P.C.V.); (M.S.G.)
| | - Mariana Soria Guerrero
- Parasitology Research Laboratory, Children Hospital of México “Federico Gómez”, Dr. Márquez 162, Col Doctores, Cuauhtémoc, Mexico City 06720, Mexico; (L.E.G.); (M.d.P.C.V.); (M.S.G.)
| | - Enedina Jiménez Cardoso
- Parasitology Research Laboratory, Children Hospital of México “Federico Gómez”, Dr. Márquez 162, Col Doctores, Cuauhtémoc, Mexico City 06720, Mexico; (L.E.G.); (M.d.P.C.V.); (M.S.G.)
| | - Marcos Meneses Mayo
- Faculty of Health Science, Anahuac Mexico University, Av. Anahuac University 46, Lomas Anáhuac, Huixquilucan, Estado de México, Huixquilucan 52786, Mexico; (M.M.M.); (S.A.I.A.)
| | - Sergio Agustín Islas Andrade
- Faculty of Health Science, Anahuac Mexico University, Av. Anahuac University 46, Lomas Anáhuac, Huixquilucan, Estado de México, Huixquilucan 52786, Mexico; (M.M.M.); (S.A.I.A.)
| |
Collapse
|
3
|
Júnior JPDL, Teixeira SC, de Souza G, Faria GV, Almeida MPO, Franco PS, Luz LC, Paschoalino M, Dos Santos NCL, de Oliveira RM, Martínez AFF, Rosini AM, Ambrosio MALV, Veneziani RCS, Bastos JK, Gomes AO, Alves RN, da Silva CV, Martins CHG, Ferro EAV, Barbosa BF. Copaifera spp. oleoresins control Trypanosoma cruzi infection in human trophoblast cells (BeWo) and placental explants. Biomed Pharmacother 2024; 179:117425. [PMID: 39265235 DOI: 10.1016/j.biopha.2024.117425] [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: 06/11/2024] [Revised: 08/25/2024] [Accepted: 09/04/2024] [Indexed: 09/14/2024] Open
Abstract
Congenital Chagas disease (CCD) is a worldwide neglected problem with significant treatment limitations. This study aimed to evaluate the potential of Copaifera spp. oleoresins (ORs) against Trypanosoma cruzi infection in trophoblast cells (BeWo lineage) and human chorionic villous explants (HCVE). The cytotoxicity of ORs was investigated using LDH and MTT assays. T. cruzi (Y strain) proliferation, invasion and reversibility were assessed in OR-treated BeWo cells, and proliferation was evaluated in OR-treated HCVE. The ultrastructure of T. cruzi trypomastigotes and amastigotes treated with ORs were analyzed by scanning and transmission electronic microscopy. ROS production in infected and treated BeWo cells and cytokines in BeWo and HCVE were measured. The ORs irreversibly decreased T. cruzi invasion, proliferation and release in BeWo cells by up to 70 %, 82 % and 80 %, respectively, and reduced parasite load in HCVE by up to 80 %. Significant structural changes in treated parasites were observed. ORs showed antioxidant capacity in BeWo cells, reducing ROS production induced by T. cruzi infection. Also, T. cruzi infection modulated the cytokine profile in both BeWo cells and HCVE; however, treatment with ORs upregulated cytokines decreased by T. cruzi infection in BeWo cells, while downregulated cytokines increased by the T. cruzi infection in HCVE. In conclusion, non-cytotoxic concentrations of Copaifera ORs demonstrated promising potential for controlling T. cruzi infection in models of the human maternal-fetal interface.
Collapse
Affiliation(s)
- Joed Pires de Lima Júnior
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Samuel Cota Teixeira
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Guilherme de Souza
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Guilherme Vieira Faria
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Marcos Paulo Oliveira Almeida
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Priscila Silva Franco
- Department of Parasitology, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Luana Carvalho Luz
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Marina Paschoalino
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Natália Carine Lima Dos Santos
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Rafael Martins de Oliveira
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Aryani Felixa Fajardo Martínez
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Alessandra Monteiro Rosini
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | | | | | - Jairo Kenupp Bastos
- School of Pharmaceutical Sciences of Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil.
| | - Angelica Oliveira Gomes
- Institute of Natural and Biological Sciences, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil.
| | - Rosiane Nascimento Alves
- Department of Agricultural and Natural Science, Universidade do Estado de Minas Gerais, Ituiutaba, MG, Brazil.
| | - Claudio Vieira da Silva
- Laboratory of Trypanosomatids, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Carlos Henrique Gomes Martins
- Laboratory of Antimicrobial Testing, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Eloisa Amália Vieira Ferro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Bellisa Freitas Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| |
Collapse
|
4
|
Boukaabar M, Oduro B, Chataa P. Congenital transmission of Chagas disease: The role of newborn therapy on the disease's dynamics. PLoS One 2024; 19:e0308136. [PMID: 39298501 DOI: 10.1371/journal.pone.0308136] [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: 02/21/2024] [Accepted: 07/15/2024] [Indexed: 09/21/2024] Open
Abstract
Chagas disease, also known as American trypanosomiasis, is caused by a protozoan blood-borne pathogen called Trypanosoma cruzi. The World Health Organization (WHO) has classified Chagas as one of 21 neglected tropical diseases present in the world and estimates that 6-7 million people are currently infected with Chagas. Congenital transmission of Chagas disease contributes to a significant amount of new infections, especially in endemic areas where 22.5% of new infections are due to congenital transmission. In this paper, we investigate congenital transmission's impact on Chagas disease dynamics through a mathematical model. Specifically, we examine how treating a proportion of infants born to infected individuals impacts the progression and spread of Chagas disease. The influence of newborn therapy on the dynamics of the model is thoroughly investigated, both theoretically and numerically. The results illustrate the importance of treating a high proportion of newborns to reduce the number of infected cases of the disease. The findings show that the therapy given to newborns is necessary but not sufficient to curb the transmission of Chagas disease, and a comprehensive approach that includes vector and vertical transmission control strategies is essential for eradicating Chagas disease. We also observed that if vector transmission can be controlled, then at least 55% of the newborns need to be treated to eliminate the disease.
Collapse
Affiliation(s)
- Meriem Boukaabar
- Department of Mathematics, Pennsylvania Western University, California, PA, United States of America
| | - Bismark Oduro
- Department of Mathematics, Pennsylvania Western University, California, PA, United States of America
| | - Paul Chataa
- Department of Mathematics, University of Cape Coast, Cape Coast, Ghana
| |
Collapse
|
5
|
Moraes FCAD, Souza MEC, Dal Moro L, Donadon IB, da Silva ER, de Souza DDSM, Burbano RMR. Prevention of congenital chagas disease by trypanocide treatment in women of reproductive age: A meta-analysis of observational studies. PLoS Negl Trop Dis 2024; 18:e0012407. [PMID: 39236037 PMCID: PMC11376591 DOI: 10.1371/journal.pntd.0012407] [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: 12/15/2023] [Accepted: 07/26/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Maternal-foetal transmission of Chagas disease (CD) affects newborns worldwide. Although Benznidazole and Nifurtimox therapies are the standard treatments, their use during pregnancy is contra-indicated. The effectiveness of trypanocidal medications in preventing congenital Chagas Disease (cCD) in the offsprings of women diagnosed with CD was highly suggested by other studies. METHODS We performed a systematic review and meta-analysis of studies evaluating the effectiveness of treatment for CD in women of childbearing age and reporting frequencies of cCD in their children. PubMed, Scopus, Web of Science, Cochrane Library, and LILACS databases were systematically searched. Statistical analysis was performed using Rstudio 4.2 using DerSimonian and Laird random-effects models. Heterogeneity was examined with the Cochran Q test and I2 statistics. A p-value of <0.05 was considered statistically significant. RESULTS Six studies were included, comprising 744 children, of whom 286 (38.4%) were born from women previously treated with Benznidazole or Nifurtimox, trypanocidal agents. The primary outcome of the proportion of children who were seropositive for cCD, confirmed by serology, was signigicantly lower among women who were previously treated with no congenital transmission registered (OR 0.05; 95% Cl 0.01-0.27; p = 0.000432; I2 = 0%). In women previously treated with trypanocidal drugs, the pooled prevalence of cCD was 0.0% (95% Cl 0-0.91%; I2 = 0%), our meta-analysis confirms the excellent effectiveness of this treatment. The prevalence of adverse events in women previously treated with antitrypanocidal therapies was 14.01% (95% CI 1.87-26.14%; I2 = 80%), Benznidazole had a higher incidence of side effects than Nifurtimox (76% vs 24%). CONCLUSION The use of trypanocidal therapy in women at reproductive age with CD is an effective strategy for the prevention of cCD, with a complete elimination of congenital transmission of Trypanosoma cruzi in treated vs untreated infected women.
Collapse
|
6
|
Rojas Panozo L, Rivera Nina S, Wehrendt DP, Casellas A, Pinto L, Mendez S, Kuo CW, Lozano DF, Ortiz L, Pinazo MJ, Picado A, Sanz S, Abril M, Gascon J, Wong S, Schijman AG, Torrico F, Alonso-Padilla J. Evaluation and validation of a PrintrLab-based LAMP assay to identify Trypanosoma cruzi in newborns in Bolivia: a proof-of-concept study. THE LANCET. MICROBE 2024; 5:100887. [PMID: 38971173 DOI: 10.1016/s2666-5247(24)00110-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 04/07/2024] [Accepted: 04/19/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND Vertical transmission of Trypanosoma cruzi represents approximately 20% of new Chagas disease cases. Early detection and treatment for women of childbearing age and newborns is a public health priority, but the lack of a simple and reliable diagnostic test remains a major barrier. We aimed to evaluate the performance of a point-of-care loop-mediated isothermal amplification (LAMP) assay for the detection of T cruzi. METHODS In this proof-of-concept study, we coupled a low-cost 3D printer repurposed for sample preparation and amplification (PrintrLab) to the Eiken T cruzi-LAMP prototype to detect vertically transmitted T cruzi, which we compared with standardised PCR and with the gold-standard algorithm (microscopy at birth and 2 months and serological study several months later). We screened pregnant women from two hospitals in the Bolivian Gran Chaco province, and those who were seropositive for T cruzi were offered the opportunity for their newborns to be enrolled in the study. Newborns were tested by microscopy, LAMP, and PCR at birth and 2 months, and by serology at 8 months. FINDINGS Between April 23 and Nov 17, 2018, 986 mothers were screened, among whom 276 were seropositive for T cruzi (28·0% prevalence, 95% CI 25·6-31·2). In total, 224 infants born to 221 seropositive mothers completed 8 months of follow-up. Congenital transmission was detected in nine of the 224 newborns (4·0% prevalence, 1·9-7·5) by direct microscopy observation, and 14 more cases were diagnosed serologically (6·3%, 3·6-10·3), accounting for an overall vertical transmission rate of 10·3% (6·6-15·0; 23 of 224). All microscopy-positive newborns were positive by PrintrLab-LAMP and by PCR, while these techniques respectively detected four and five extra positive cases among the remaining 215 microscopy-negative newborns. INTERPRETATION The PrintrLab-LAMP yielded a higher sensitivity than microscopy-based analysis. Considering the simpler use and expected lower cost of LAMP compared with PCR, our findings encourage its evaluation in a larger study over a wider geographical area. FUNDING Inter-American Development Bank.
Collapse
Affiliation(s)
- Lizeth Rojas Panozo
- Fundación Ciencias y Estudios Aplicados para el Desarrollo en Salud y Medio Ambiente (CEADES), Cochabamba, Bolivia
| | - Silvia Rivera Nina
- Fundación Ciencias y Estudios Aplicados para el Desarrollo en Salud y Medio Ambiente (CEADES), Cochabamba, Bolivia
| | - Diana P Wehrendt
- Laboratorio de Biología Molecular de La Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Buenos Aires, Argentina
| | - Aina Casellas
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic - University of Barcelona, Barcelona, Spain
| | - Lilian Pinto
- Fundación Ciencias y Estudios Aplicados para el Desarrollo en Salud y Medio Ambiente (CEADES), Cochabamba, Bolivia
| | - Susana Mendez
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic - University of Barcelona, Barcelona, Spain
| | | | - Daniel F Lozano
- Fundación Ciencias y Estudios Aplicados para el Desarrollo en Salud y Medio Ambiente (CEADES), Cochabamba, Bolivia; Universidad Mayor de San Simón, Cochabamba, Bolivia
| | - Lourdes Ortiz
- Plataforma de Chagas Tarija - Universidad Autónoma Juan Misael Saracho, Tarija, Bolivia
| | - Maria-Jesus Pinazo
- Drugs for Neglected Diseases Initiative (DNDi), Rio de Janeiro, Brazil; CIBER de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Albert Picado
- Foundation for Innovative Diagnostics (FIND), Geneva, Switzerland
| | - Sergi Sanz
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic - University of Barcelona, Barcelona, Spain
| | | | - Joaquim Gascon
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic - University of Barcelona, Barcelona, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | | | - Alejandro G Schijman
- Laboratorio de Biología Molecular de La Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Buenos Aires, Argentina
| | - Faustino Torrico
- Fundación Ciencias y Estudios Aplicados para el Desarrollo en Salud y Medio Ambiente (CEADES), Cochabamba, Bolivia; Universidad Mayor de San Simón, Cochabamba, Bolivia
| | - Julio Alonso-Padilla
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic - University of Barcelona, Barcelona, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain.
| |
Collapse
|
7
|
Apodaca S, Di Salvatore M, Muñoz-Calderón A, Curto MDLÁ, Longhi SA, Schijman AG. Novel 3D human trophoblast culture to explore T. cruzi infection in the placenta. Front Cell Infect Microbiol 2024; 14:1433424. [PMID: 39165920 PMCID: PMC11333438 DOI: 10.3389/fcimb.2024.1433424] [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: 05/15/2024] [Accepted: 07/08/2024] [Indexed: 08/22/2024] Open
Abstract
Introduction Human trophoblastic cell lines, such as BeWo, are commonly used in 2D models to study placental Trypanosoma cruzi infections. However, these models do not accurately represent natural infections. Three-dimensional (3D) microtissue cultures offer a more physiologically relevant in vitro model, mimicking tissue microarchitecture and providing an environment closer to natural infections. These 3D cultures exhibit functions such as cell proliferation, differentiation, morphogenesis, and gene expression that resemble in vivo conditions. Methods We developed a 3D culture model using the human trophoblastic cell line BeWo and nonadherent agarose molds from the MicroTissues® 3D Petri Dish® system. Both small (12-256) and large (12-81) models were tested with varying initial cell numbers. We measured the diameter of the 3D cultures and evaluated cell viability using Trypan Blue dye. Trophoblast functionality was assessed by measuring β-hCG production via ELISA. Cell fusion was evaluated using confocal microscopy, with Phalloidin or ZO-1 marking cell edges and DAPI staining nuclei. T. cruzi infection was assessed by microscopy and quantitative PCR, targeting the EF1-α gene for T. cruzi and GAPDH for BeWo cells, using three parasite strains: VD (isolated from a congenital Chagas disease infant and classified as Tc VI), and K98 and Pan4 (unrelated to congenital infection and classified as Tc I). Results Seeding 1000 BeWo cells per microwell in the large model resulted in comparable cellular viability to 2D cultures, with a theoretical diameter of 408.68 ± 12.65 μm observed at 5 days. Functionality, assessed through β-hCG production, exceeded levels in 2D cultures at both 3 and 5 days. T. cruzi infection was confirmed by qPCR and microscopy, showing parasite presence inside the cells for all three tested strains. The distribution and progression of the infection varied with each strain. Discussion This innovative 3D model offers a simple yet effective approach for generating viable and functional cultures susceptible to T. cruzi infection, presenting significant potential for studying the placental microenvironment.
Collapse
Affiliation(s)
| | | | | | | | | | - Alejandro G. Schijman
- Laboratorio de Biología Molecular de la Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
8
|
Schijman AG, Alonso-Padilla J, Britto C, Herrera Bernal CP. Retrospect, advances and challenges in Chagas disease diagnosis: a comprehensive review. LANCET REGIONAL HEALTH. AMERICAS 2024; 36:100821. [PMID: 39006126 PMCID: PMC11246061 DOI: 10.1016/j.lana.2024.100821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/29/2024] [Accepted: 06/05/2024] [Indexed: 07/16/2024]
Abstract
Chagas disease, caused by Trypanosoma cruzi, affects millions worldwide. The 2030 WHO roadmap aims to eliminate it as a public health concern, emphasising the need for timely diagnosis to enhance treatment access. Current diagnostic algorithms, which rely on multiple tests, have prolonged turnaround times. This proves particularly problematic in resource-limited settings. Addressing this issue necessitates the validation and adoption of innovative tools. We explore recent developments in Chagas disease diagnosis, reviewing historical context and advancements. Despite progress, challenges persist. This article contributes to the understanding of current and future directions in this neglected healthcare area. Parasitological methods are simple but exhibit low sensitivity and require supplementary tests. Molecular methods, with automation potential, allow quantification and higher throughput. Serological tools show good performance but struggle with parasite antigenic diversity. Prioritising point-of-care tests is crucial for widespread accessibility and could offer a strategy to control disease impact. Ultimately, balancing achievements and ongoing obstacles is essential for comprehensive progress.
Collapse
Affiliation(s)
- Alejandro Gabriel Schijman
- Laboratorio de Biología Molecular de la Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr Héctor Torres", INGEBI- CONICET, Vuelta de Obligado 2490, Buenos Aires, 1428 ADN, Argentina
| | - Julio Alonso-Padilla
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic - University of Barcelona, Carrer Rosselló 149, 08036, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Constança Britto
- Fundação Oswaldo Cruz, Fiocruz, Instituto Oswaldo Cruz, Laboratory of Molecular Biology and Endemic Diseases, Avenida Brasil 4365, Manguinhos, Rio de Janeiro, 21045-900, Brazil
| | | |
Collapse
|
9
|
Silvestrini MMA, Alessio GD, Frias BED, Sales Júnior PA, Araújo MSS, Silvestrini CMA, Brito Alvim de Melo GE, Martins-Filho OA, Teixeira-Carvalho A, Martins HR. New insights into Trypanosoma cruzi genetic diversity, and its influence on parasite biology and clinical outcomes. Front Immunol 2024; 15:1342431. [PMID: 38655255 PMCID: PMC11035809 DOI: 10.3389/fimmu.2024.1342431] [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: 11/21/2023] [Accepted: 02/26/2024] [Indexed: 04/26/2024] Open
Abstract
Chagas disease, caused by Trypanosoma cruzi, remains a serious public health problem worldwide. The parasite was subdivided into six distinct genetic groups, called "discrete typing units" (DTUs), from TcI to TcVI. Several studies have indicated that the heterogeneity of T. cruzi species directly affects the diversity of clinical manifestations of Chagas disease, control, diagnosis performance, and susceptibility to treatment. Thus, this review aims to describe how T. cruzi genetic diversity influences the biology of the parasite and/or clinical parameters in humans. Regarding the geographic dispersion of T. cruzi, evident differences were observed in the distribution of DTUs in distinct areas. For example, TcII is the main DTU detected in Brazilian patients from the central and southeastern regions, where there are also registers of TcVI as a secondary T. cruzi DTU. An important aspect observed in previous studies is that the genetic variability of T. cruzi can impact parasite infectivity, reproduction, and differentiation in the vectors. It has been proposed that T. cruzi DTU influences the host immune response and affects disease progression. Genetic aspects of the parasite play an important role in determining which host tissues will be infected, thus heavily influencing Chagas disease's pathogenesis. Several teams have investigated the correlation between T. cruzi DTU and the reactivation of Chagas disease. In agreement with these data, it is reasonable to suppose that the immunological condition of the patient, whether or not associated with the reactivation of the T. cruzi infection and the parasite strain, may have an important role in the pathogenesis of Chagas disease. In this context, understanding the genetics of T. cruzi and its biological and clinical implications will provide new knowledge that may contribute to additional strategies in the diagnosis and clinical outcome follow-up of patients with Chagas disease, in addition to the reactivation of immunocompromised patients infected with T. cruzi.
Collapse
Affiliation(s)
| | - Glaucia Diniz Alessio
- Integrated Biomarker Research Group, René Rachou Institute, Fiocruz Minas, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | - Bruna Estefânia Diniz Frias
- Integrated Biomarker Research Group, René Rachou Institute, Fiocruz Minas, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | - Policarpo Ademar Sales Júnior
- Integrated Biomarker Research Group, René Rachou Institute, Fiocruz Minas, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | - Márcio Sobreira Silva Araújo
- Integrated Biomarker Research Group, René Rachou Institute, Fiocruz Minas, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | | | | | - Olindo Assis Martins-Filho
- Integrated Biomarker Research Group, René Rachou Institute, Fiocruz Minas, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | - Andréa Teixeira-Carvalho
- Integrated Biomarker Research Group, René Rachou Institute, Fiocruz Minas, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | - Helen Rodrigues Martins
- Department of Pharmacy, Federal University of the Jequitinhonha and Mucuri Valleys, Diamantina, Minas Gerais, Brazil
| |
Collapse
|
10
|
de Sousa AS, Vermeij D, Ramos AN, Luquetti AO. Chagas disease. Lancet 2024; 403:203-218. [PMID: 38071985 DOI: 10.1016/s0140-6736(23)01787-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 08/08/2023] [Accepted: 08/21/2023] [Indexed: 01/15/2024]
Abstract
Chagas disease persists as a global public health problem due to the high morbidity and mortality burden. Despite the possibility of a cure and advances in transmission control, epidemiological transformations, such as urbanisation and globalisation, and the emerging importance of oral and vertical transmission mean that Chagas disease should be considered an emerging disease, with new cases occurring worldwide. Important barriers to diagnosis, treatment, and care remain, resulting in repressed numbers of reported cases, which in turn leads to inadequate public policies. The validation of new diagnostic tools and treatment options is needed, as existing tools pose serious limitations to access to health care. Integrated models of surveillance, with community and intersectional participation, embedded in the concept of One Health, are essential for control. In addition, mitigation strategies for the main social determinants of health, including difficulties imposed by migration, are important to improve access to comprehensive health care in a globalised scenario.
Collapse
Affiliation(s)
- Andréa Silvestre de Sousa
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Department of Internal Medicine, School of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Debbie Vermeij
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Alberto Novaes Ramos
- Department of Community Health, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Alejandro O Luquetti
- Center of Studies for Chagas Disease, Hospital das Clínicas, Federal University of Goiás, Goiânia, Brazil
| |
Collapse
|
11
|
Rios LE, Lokugamage N, Choudhuri S, Chowdhury IH, Garg NJ. Subunit nanovaccine elicited T cell functional activation controls Trypanosoma cruzi mediated maternal and placental tissue damage and improves pregnancy outcomes in mice. NPJ Vaccines 2023; 8:188. [PMID: 38104118 PMCID: PMC10725459 DOI: 10.1038/s41541-023-00782-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 11/10/2023] [Indexed: 12/19/2023] Open
Abstract
This study investigated a candidate vaccine effect against maternal Trypanosoma cruzi (Tc) infection and improved pregnancy outcomes. For this, TcG2 and TcG4 were cloned in a nanoplasmid optimized for delivery, antigen expression, and regulatory compliance (nano2/4 vaccine). Female C57BL/6 mice were immunized with nano2/4, infected (Tc SylvioX10), and mated 7-days post-infection to enable fetal development during the maternal acute parasitemia phase. Females were euthanized at E12-E17 (gestation) days. Splenic and placental T-cell responses were monitored by flow cytometry. Maternal and placental/fetal tissues were examined for parasites by qPCR and inflammatory infiltrate by histology. Controls included age/immunization-matched non-pregnant females. Nano2/4 exhibited no toxicity and elicited protective IgG2a/IgG1 response in mice. Nano2/4 signaled a splenic expansion of functionally active CD4+ effector/effector memory (Tem) and central memory (Tcm) cells in pregnant mice. Upon challenge infection, nano2/4 increased the splenic CD4+ and CD8+T cells in all mice and increased the proliferation of CD4+Tem, CD4+Tcm, and CD8+Tcm subsets producing IFNγ and cytolytic molecules (PRF1, GZB) in pregnant mice. A balanced serum cytokines/chemokines response and placental immune characteristics indicated that pregnancy prevented the overwhelming damaging immune response in mice. Importantly, pregnancy itself resulted in a significant reduction of parasites in maternal and fetal tissues. Nano2/4 was effective in arresting the Tc-induced tissue inflammatory infiltrate, necrosis, and fibrosis in maternal and placental tissues and improving maternal fertility, placental efficiency, and fetal survival. In conclusion, we show that maternal nano2/4 vaccination is beneficial in controlling the adverse effects of Tc infection on maternal health, fetal survival, and pregnancy outcomes.
Collapse
Affiliation(s)
- Lizette Elaine Rios
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
- Department of Biochemistry and Molecular Biology, UTMB, Galveston, TX, USA
| | - Nandadeva Lokugamage
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Subhadip Choudhuri
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Imran Hussain Chowdhury
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Nisha Jain Garg
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, TX, USA.
- Institute for Human Infections and Immunity (IHII), UTMB, Galveston, TX, USA.
- Sealy Institute for Vaccine Sciences (SIVS), UTMB, Galveston, TX, USA.
| |
Collapse
|
12
|
Guerrero-Muñoz J, Medina L, Castillo C, Liempi A, Fernández-Moya A, Araneda S, Ortega Y, Rojas-Pirela M, Maya JD, Kemmerling U. MicroRNA-512-3p mediates Trypanosoma cruzi-induced apoptosis during ex vivo infection of human placental explants. Placenta 2023; 143:117-123. [PMID: 37898020 DOI: 10.1016/j.placenta.2023.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 09/25/2023] [Accepted: 10/16/2023] [Indexed: 10/30/2023]
Abstract
INTRODUCTION Upon infection, Trypanosoma cruzi, a protozoan parasite, crosses the placental barrier and causes congenital Chagas disease. Ex vivo infection of human placental explants (HPEs) with the parasite induces apoptotic cell death. This cellular process involves changes in gene expression, which are partially regulated by miRNAs. In this study, we investigated the role of miR-512-3p, a highly expressed miRNA in the placenta, in parasite-induced apoptosis. METHODS HPE cells were transfected with antagomirs or mimics of miR-512-3p and subsequently challenged with the parasite. The expression levels of miR-512-3p, caspase 3, caspase 8, and Livin were measured using RT-qPCR, and apoptotic cell death was analyzed based on caspase activity and DNA fragmentation assays. RESULTS Targeted inhibition of miR-512-3p effectively prevented parasite-induced expression and enzymatic activity of caspase 3 and caspase 8. However, it did not completely prevent DNA fragmentation, indicating the involvement of other factors in this process. Furthermore, the findings suggest that Livin may be regulated by miR-512-3p. DISCUSSION Our findings suggest that miR-512-3p modulates parasite-induced apoptosis in the trophoblast. By understanding the mechanisms involved in this process, we can gain insights into the pathogenesis of congenital Chagas disease and develop targeted therapeutic strategies.
Collapse
Affiliation(s)
- Jesús Guerrero-Muñoz
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Lisvaneth Medina
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Christian Castillo
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Núcleo de Investigación Aplicada en Ciencias Veterinarias y Agronómicas, Facultad de Medicina Veterinaria y Agronomía, Universidad de Las Américas, Chile
| | - Ana Liempi
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Alejandro Fernández-Moya
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Sebastian Araneda
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Departamento de Patología y Medicina oral, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Yessica Ortega
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Facultad de Farmacia y Bioanálisis, Universidad de Los Andes, Mérida, Venezuela
| | - Maura Rojas-Pirela
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Juan Diego Maya
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Ulrike Kemmerling
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
13
|
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
|
14
|
Gabaldón-Figueira JC, Skjefte M, Longhi S, Escabia E, García LJ, Ros-Lucas A, Martínez-Peinado N, Muñoz-Calderón A, Gascón J, Schijman AG, Alonso-Padilla J. Practical diagnostic algorithms for Chagas disease: a focus on low resource settings. Expert Rev Anti Infect Ther 2023; 21:1287-1299. [PMID: 37933443 DOI: 10.1080/14787210.2023.2279110] [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: 08/08/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023]
Abstract
INTRODUCTION Chagas disease, caused by parasite Trypanosoma cruzi, is the most important neglected tropical disease in the Americas. Two drugs are available for treatment, but access to them is challenging, in part due to complex diagnostic algorithms. These are stage-dependent, involve multiple tests, and are ill-adapted to the reality of vast areas where the disease is endemic. Molecular and serologic tools are used to detect acute and chronic infections, with the performance of the latter showing geographic differences. Breakthroughs in the development of new diagnostic tools include the validation of a loop-mediated isothermal amplification assay for acute infections (T. cruzi-LAMP), and the regional validation of several rapid diagnostic tests (RDTs) for chronic infection, which simplify testing in resource-limited settings. The literature search was carried out in the MEDLINE database until 1 August 2023. AREAS COVERED This review outlines existing algorithms, and proposes new ones focused on point-of-care testing. EXPERT OPINION Integrating point-of-care testing into existing diagnostic algorithms in certain endemic areas will increase access to timely diagnosis and treatment. However, additional research is needed to validate the use of these techniques across a wider geography, and to better understand the cost-effectiveness of their large-scale implementation.
Collapse
Affiliation(s)
| | - Malia Skjefte
- Population Services International (PSI), Washington, MA, USA
| | - Silvia Longhi
- Laboratorio de Biología Molecular de la Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr Héctor Torres", INGEBI-CONICET, Buenos Aires, Argentina
| | - Elisa Escabia
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic-University of Barcelona, Barcelona, Spain
| | - Lady Juliette García
- Laboratorio de Biología Molecular de la Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr Héctor Torres", INGEBI-CONICET, Buenos Aires, Argentina
| | - Albert Ros-Lucas
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic-University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
| | - Nieves Martínez-Peinado
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic-University of Barcelona, Barcelona, Spain
| | - Arturo Muñoz-Calderón
- Laboratorio de Biología Molecular de la Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr Héctor Torres", INGEBI-CONICET, Buenos Aires, Argentina
| | - Joaquim Gascón
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic-University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
| | - Alejandro Gabriel Schijman
- Laboratorio de Biología Molecular de la Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr Héctor Torres", INGEBI-CONICET, Buenos Aires, Argentina
| | - Julio Alonso-Padilla
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic-University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
| |
Collapse
|
15
|
Marin-Neto JA, Rassi A, Oliveira GMM, Correia LCL, Ramos Júnior AN, Luquetti AO, Hasslocher-Moreno AM, Sousa ASD, Paola AAVD, Sousa ACS, Ribeiro ALP, Correia Filho D, Souza DDSMD, Cunha-Neto E, Ramires FJA, Bacal F, Nunes MDCP, Martinelli Filho M, Scanavacca MI, Saraiva RM, Oliveira Júnior WAD, Lorga-Filho AM, Guimarães ADJBDA, Braga ALL, Oliveira ASD, Sarabanda AVL, Pinto AYDN, Carmo AALD, Schmidt A, Costa ARD, Ianni BM, Markman Filho B, Rochitte CE, Macêdo CT, Mady C, Chevillard C, Virgens CMBD, Castro CND, Britto CFDPDC, Pisani C, Rassi DDC, Sobral Filho DC, Almeida DRD, Bocchi EA, Mesquita ET, Mendes FDSNS, Gondim FTP, Silva GMSD, Peixoto GDL, Lima GGD, Veloso HH, Moreira HT, Lopes HB, Pinto IMF, Ferreira JMBB, Nunes JPS, Barreto-Filho JAS, Saraiva JFK, Lannes-Vieira J, Oliveira JLM, Armaganijan LV, Martins LC, Sangenis LHC, Barbosa MPT, Almeida-Santos MA, Simões MV, Yasuda MAS, Moreira MDCV, Higuchi MDL, Monteiro MRDCC, Mediano MFF, Lima MM, Oliveira MTD, Romano MMD, Araujo NNSLD, Medeiros PDTJ, Alves RV, Teixeira RA, Pedrosa RC, Aras Junior R, Torres RM, Povoa RMDS, Rassi SG, Alves SMM, Tavares SBDN, Palmeira SL, Silva Júnior TLD, Rodrigues TDR, Madrini Junior V, Brant VMDC, Dutra WO, Dias JCP. SBC Guideline on the Diagnosis and Treatment of Patients with Cardiomyopathy of Chagas Disease - 2023. Arq Bras Cardiol 2023; 120:e20230269. [PMID: 37377258 PMCID: PMC10344417 DOI: 10.36660/abc.20230269] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023] Open
Affiliation(s)
- José Antonio Marin-Neto
- Universidade de São Paulo , Faculdade de Medicina de Ribeirão Preto , Ribeirão Preto , SP - Brasil
| | - Anis Rassi
- Hospital do Coração Anis Rassi , Goiânia , GO - Brasil
| | | | | | | | - Alejandro Ostermayer Luquetti
- Centro de Estudos da Doença de Chagas , Hospital das Clínicas da Universidade Federal de Goiás , Goiânia , GO - Brasil
| | | | - Andréa Silvestre de Sousa
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| | | | - Antônio Carlos Sobral Sousa
- Universidade Federal de Sergipe , São Cristóvão , SE - Brasil
- Hospital São Lucas , Rede D`Or São Luiz , Aracaju , SE - Brasil
| | | | | | | | - Edecio Cunha-Neto
- Universidade de São Paulo , Faculdade de Medicina da Universidade, São Paulo , SP - Brasil
| | - Felix Jose Alvarez Ramires
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Fernando Bacal
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | - Martino Martinelli Filho
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Maurício Ibrahim Scanavacca
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Roberto Magalhães Saraiva
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| | | | - Adalberto Menezes Lorga-Filho
- Instituto de Moléstias Cardiovasculares , São José do Rio Preto , SP - Brasil
- Hospital de Base de Rio Preto , São José do Rio Preto , SP - Brasil
| | | | | | - Adriana Sarmento de Oliveira
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | - Ana Yecê das Neves Pinto
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| | | | - Andre Schmidt
- Universidade de São Paulo , Faculdade de Medicina de Ribeirão Preto , Ribeirão Preto , SP - Brasil
| | - Andréa Rodrigues da Costa
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| | - Barbara Maria Ianni
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | - Carlos Eduardo Rochitte
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
- Hcor , Associação Beneficente Síria , São Paulo , SP - Brasil
| | | | - Charles Mady
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Christophe Chevillard
- Institut National de la Santé Et de la Recherche Médicale (INSERM), Marselha - França
| | | | | | | | - Cristiano Pisani
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | | | | | - Edimar Alcides Bocchi
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Evandro Tinoco Mesquita
- Hospital Universitário Antônio Pedro da Faculdade Federal Fluminense , Niterói , RJ - Brasil
| | | | | | | | | | | | - Henrique Horta Veloso
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| | - Henrique Turin Moreira
- Hospital das Clínicas , Faculdade de Medicina de Ribeirão Preto , Universidade de São Paulo , Ribeirão Preto , SP - Brasil
| | | | | | | | - João Paulo Silva Nunes
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
- Fundação Zerbini, Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | | | | | | | | | - Luiz Cláudio Martins
- Universidade Estadual de Campinas , Faculdade de Ciências Médicas , Campinas , SP - Brasil
| | | | | | | | - Marcos Vinicius Simões
- Universidade de São Paulo , Faculdade de Medicina de Ribeirão Preto , Ribeirão Preto , SP - Brasil
| | | | | | - Maria de Lourdes Higuchi
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | - Mauro Felippe Felix Mediano
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
- Instituto Nacional de Cardiologia (INC), Rio de Janeiro, RJ - Brasil
| | - Mayara Maia Lima
- Secretaria de Vigilância em Saúde , Ministério da Saúde , Brasília , DF - Brasil
| | | | | | | | | | - Renato Vieira Alves
- Instituto René Rachou , Fundação Oswaldo Cruz , Belo Horizonte , MG - Brasil
| | - Ricardo Alkmim Teixeira
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Roberto Coury Pedrosa
- Hospital Universitário Clementino Fraga Filho , Instituto do Coração Edson Saad - Universidade Federal do Rio de Janeiro , RJ - Brasil
| | | | | | | | | | - Silvia Marinho Martins Alves
- Ambulatório de Doença de Chagas e Insuficiência Cardíaca do Pronto Socorro Cardiológico Universitário da Universidade de Pernambuco (PROCAPE/UPE), Recife , PE - Brasil
| | | | - Swamy Lima Palmeira
- Secretaria de Vigilância em Saúde , Ministério da Saúde , Brasília , DF - Brasil
| | | | | | - Vagner Madrini Junior
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | | | - João Carlos Pinto Dias
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| |
Collapse
|
16
|
Rios LE, Lokugamage N, Garg NJ. Effects of Acute and Chronic Trypanosoma cruzi Infection on Pregnancy Outcomes in Mice: Parasite Transmission, Mortality, Delayed Growth, and Organ Damage in Pups. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:313-331. [PMID: 36565805 PMCID: PMC10013038 DOI: 10.1016/j.ajpath.2022.11.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/20/2022] [Accepted: 11/30/2022] [Indexed: 12/22/2022]
Abstract
Chagas disease is caused by Trypanosoma cruzi. This study aimed to determine the effects of T. cruzi infection on fertility rate and health of the newborn pups in pregnant mice. Female mice were challenged with T. cruzi and mated at 21 days (acute parasitemic phase) or 90 days (chronic parasite persistence phase) after infection. Pups were examined for growth up to 20 days after birth; and parasite burden in brain, heart, skeletal muscle, and intestine was measured by real-time quantitative PCR. The inflammatory infiltrate, necrosis, and fibrosis in pups' heart and brain tissues were evaluated by histology. T. cruzi infection in dams delayed the onset of pregnancy, decreased the fertility rate, and led to vertical transmission of parasite to the pups. Furthermore, infected dams delivered pups that exhibited decreased survival rate, decreased birth weight, and decreased growth rate. Significantly increased inflammation, necrosis, and fibrosis of cardiac and brain tissues were noted in pups born to infected dams. Initial challenge with higher parasite dose had more detrimental effects on fertility rate and pups' health in both acutely and chronically infected dams. In conclusion, mice offer a promising model to evaluate the efficacy of new vaccines and therapeutic drugs in controlling the acute and chronic maternal T. cruzi infection and congenital transmission to newborns, and in improving the fertility rate and pups' health outcomes.
Collapse
Affiliation(s)
- Lizette E Rios
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas; Department of Biochemistry, Cellular and Molecular Biology, University of Texas Medical Branch, Galveston, Texas
| | - Nandadeva Lokugamage
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Nisha J Garg
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas.
| |
Collapse
|
17
|
Matthews S, Tannis A, Puchner KP, Bottazzi ME, Cafferata ML, Comandé D, Buekens P. Estimation of the morbidity and mortality of congenital Chagas disease: A systematic review and meta-analysis. PLoS Negl Trop Dis 2022; 16:e0010376. [PMID: 36342961 PMCID: PMC9671465 DOI: 10.1371/journal.pntd.0010376] [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: 03/31/2022] [Revised: 11/17/2022] [Accepted: 10/21/2022] [Indexed: 11/09/2022] Open
Abstract
Chagas disease is caused by the parasite Trypanosoma cruzi which can be transmitted from mother to baby during pregnancy. There is no consensus on the proportion of infected infants with clinical signs of congenital Chagas disease (cCD). The objective of this systematic review is to determine the burden of cCD. Articles from journal inception to 2020 reporting morbidity and mortality associated with cCD were retrieved from academic search databases. Observational studies, randomized-control trials, and studies of babies diagnosed with cCD were included. Studies were excluded if they were case reports or series, without original data, case-control without cCD incidence estimates, and/or did not report number of participants. Two reviewers screened articles for inclusion. To determine pooled proportion of infants with cCD with clinical signs, individual clinical signs, and case-fatality, random effects meta-analysis was performed. We identified 4,531 records and reviewed 4,301, including 47 articles in the narrative summary and analysis. Twenty-eight percent of cCD infants showed clinical signs (95% confidence interval (CI) = 19.0%, 38.5%) and 2.2% of infants died (95% CI = 1.3%, 3.5%). The proportion of infected infants with hepatosplenomegaly was 12.5%, preterm birth 6.0%, low birth weight 5.8%, anemia 4.9%, and jaundice 4.7%. Although most studies did not include a comparison group of non-infected infants, the proportion of infants with cCD with clinical signs at birth are comparable to those with congenital toxoplasmosis (10.0%-30.0%) and congenital cytomegalovirus (10.0%-15.0%). We conclude that cCD burden appears significant, but more studies comparing infected mother-infant dyads to non-infected ones are needed to determine an association of this burden to cCD.
Collapse
Affiliation(s)
- Sarah Matthews
- Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - Ayzsa Tannis
- Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | | | - Maria Elena Bottazzi
- National School of Tropical Medicine, Department of Pediatrics, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
- Texas Children’s Hospital Center for Vaccine Development, Houston, Texas, United States of America
| | - Maria Luisa Cafferata
- Instituto de Efectividad Clínica y Sanitaria, Buenos Aires, Argentina
- Unidad de Investigación Clínica y Epidemiológica Montevideo (UNICEM), Montevideo, Uruguay
| | - Daniel Comandé
- Instituto de Efectividad Clínica y Sanitaria, Buenos Aires, Argentina
| | - Pierre Buekens
- Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| |
Collapse
|
18
|
Avalos-Borges EE, Rios LE, Jiménez-Coello M, Ortega-Pacheco A, Garg NJ. Animal Models of Trypanosoma cruzi Congenital Transmission. Pathogens 2022; 11:1172. [PMID: 36297229 PMCID: PMC9611621 DOI: 10.3390/pathogens11101172] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/03/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Chagas disease, initiated by the etiological agent Trypanosoma cruzi, is an endemic infection in the American continent. Although vectorial transmission of T. cruzi is recognized as the main mode of infection, other routes such as congenital and blood transfusion are also documented as important methods of transmission. T. cruzi maternal-fetal transmission has been recorded in humans and examined by some investigators in naturally and experimentally infected mammals. Dogs are recognized as the major reservoir host in maintaining the domestic transmission of T. cruzi; however, the importance of congenital transmission in preserving the infection cycle in dogs has not been studied in detail. In this article, we reviewed the current knowledge of congenital transmission of T. cruzi in humans and compared the placental architecture of humans and different animals with particular attention to rodents, dogs, and non-human primates that have been used as experimental models of T. cruzi infection, congenital transmission, and Chagas disease pathogenesis. The placentas of humans and animals have some similar and dissimilar characteristics that should inform the study design and interpretation of results when evaluating the efficacy of new anti-parasite drugs and therapies against congenital infection.
Collapse
Affiliation(s)
- Eduardo E. Avalos-Borges
- Departamento de Salud Animal y Medicina Preventiva, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Yucatan, Merida 97000, Yucatan, Mexico
| | - Lizette E. Rios
- Department of Microbiology & Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-1070, USA
| | - Matilde Jiménez-Coello
- Departamento de Salud Animal y Medicina Preventiva, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Yucatan, Merida 97000, Yucatan, Mexico
| | - Antonio Ortega-Pacheco
- Departamento de Salud Animal y Medicina Preventiva, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Yucatan, Merida 97000, Yucatan, Mexico
| | - Nisha J. Garg
- Department of Microbiology & Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-1070, USA
| |
Collapse
|
19
|
de Arias AR, Monroy C, Guhl F, Sosa-Estani S, Santos WS, Abad-Franch F. Chagas disease control-surveillance in the Americas: the multinational initiatives and the practical impossibility of interrupting vector-borne Trypanosoma cruzi transmission. Mem Inst Oswaldo Cruz 2022; 117:e210130. [PMID: 35830010 PMCID: PMC9261920 DOI: 10.1590/0074-02760210130] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 11/21/2022] Open
Abstract
Chagas disease (CD) still imposes a heavy burden on most Latin American countries. Vector-borne and mother-to-child transmission cause several thousand new infections per year, and at least 5 million people carry Trypanosoma cruzi. Access to diagnosis and medical care, however, is far from universal. Starting in the 1990s, CD-endemic countries and the Pan American Health Organization-World Health Organization (PAHO-WHO) launched a series of multinational initiatives for CD control-surveillance. An overview of the initiatives’ aims, achievements, and challenges reveals some key common themes that we discuss here in the context of the WHO 2030 goals for CD. Transmission of T. cruzi via blood transfusion and organ transplantation is effectively under control. T. cruzi, however, is a zoonotic pathogen with 100+ vector species widely spread across the Americas; interrupting vector-borne transmission seems therefore unfeasible. Stronger surveillance systems are, and will continue to be, needed to monitor and control CD. Prevention of vertical transmission demands boosting current efforts to screen pregnant and childbearing-aged women. Finally, integral patient care is a critical unmet need in most countries. The decades-long experience of the initiatives, in sum, hints at the practical impossibility of interrupting vector-borne T. cruzi transmission in the Americas. The concept of disease control seems to provide a more realistic description of what can in effect be achieved by 2030.
Collapse
Affiliation(s)
| | - Carlota Monroy
- Universidad de San Carlos, Laboratorio de Entomología y Parasitología Aplicadas, Ciudad de Guatemala, Guatemala
| | - Felipe Guhl
- Universidad de los Andes, Facultad de Ciencias, Centro de Investigaciones en Microbiología y Parasitología Tropical, Bogotá, Colombia
| | - Sergio Sosa-Estani
- Drugs for Neglected Diseases initiative Latin America, Rio de Janeiro, RJ, Brasil.,Centro de Investigaciones en Epidemiología y Salud Pública, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Walter Souza Santos
- Ministério da Saúde, Secretaria de Vigilância em Saúde, Instituto Evandro Chagas, Laboratório de Epidemiologia das Leishmanioses, Ananindeua, PA, Brasil
| | - Fernando Abad-Franch
- Universidade de Brasília, Faculdade de Medicina, Núcleo de Medicina Tropical, Brasília, DF, Brasil
| |
Collapse
|
20
|
Schijman AG, Alonso-Padilla J, Longhi SA, Picado A. Parasitological, serological and molecular diagnosis of acute and chronic Chagas disease: from field to laboratory. Mem Inst Oswaldo Cruz 2022; 117:e200444. [PMID: 35613155 PMCID: PMC9164950 DOI: 10.1590/0074-02760200444] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 01/13/2021] [Indexed: 01/05/2023] Open
Abstract
There is no consensus on the diagnostic algorithms for many scenarios of Trypanosoma cruzi infection, which hinders the establishment of governmental guidelines in endemic and non-endemic countries. In the acute phase, parasitological methods are currently employed, and standardised surrogate molecular tests are being introduced to provide higher sensitivity and less operator-dependence. In the chronic phase, IgG-based serological assays are currently used, but if a single assay does not reach the required accuracy, PAHO/WHO recommends at least two immunological tests with different technical principles. Specific algorithms are applied to diagnose congenital infection, screen blood and organ donors or conduct epidemiological surveys. Detecting Chagas disease reactivation in immunosuppressed individuals is an area of increasing interest. Due to its neglect, enhancing access to diagnosis of patients at risk of suffering T. cruzi infection should be a priority at national and regional levels.
Collapse
Affiliation(s)
- Alejandro Gabriel Schijman
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular Dr Hector Torres, CONICET, Laboratorio de Biología Molecular de la Enfermedad de Chagas, Ciudad de Buenos Aires, Argentina
| | - Julio Alonso-Padilla
- Barcelona Institute for Global Health, University of Barcelona, Hospital Clinic, Barcelona, Spain
| | - Silvia Andrea Longhi
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular Dr Hector Torres, CONICET, Laboratorio de Biología Molecular de la Enfermedad de Chagas, Ciudad de Buenos Aires, Argentina
| | - Albert Picado
- Foundation for Innovative New Diagnostics, Geneva, Switzerland
| |
Collapse
|
21
|
Gorla DE, Xiao-Nong Z, Diotaiuti L, Khoa PT, Waleckx E, de Souza RDCM, Qin L, Lam TX, Freilij H. Different profiles and epidemiological scenarios: past, present and future. Mem Inst Oswaldo Cruz 2022; 117:e200409. [PMID: 35613154 PMCID: PMC9126320 DOI: 10.1590/0074-02760200409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 01/13/2021] [Indexed: 11/22/2022] Open
Abstract
The multiplicity of epidemiological scenarios shown by Chagas Disease, derived from multiple transmission routes of the aetiological agent, occurring on multiple geo-ecobiosocial settings determines the complexity of the disease and reveal the difficulties for its control. From the first description of the link between the parasite, the vector and its domestic habitat and the disease that Carlos Chagas made in 1909, the epidemiological scenarios of the American Trypanosomiasis has shown a dynamic increasing complexity. These scenarios changed with time and geography because of new understandings of the disease from multiple studies, because of policies change at the national and international levels and because human movements brought the parasite and vectors to new geographies. Paradigms that seemed solid at a time were broken down, and we learnt about the global dispersion of Trypanosoma cruzi infection, the multiplicity of transmission routes, that the infection can be cured, and that triatomines are not only a health threat in Latin America. We consider the multiple epidemiological scenarios through the different T. cruzi transmission routes, with or without the participation of a Triatominae vector. We then consider the scenario of regions with vectors without the parasite, to finish with the consideration of future prospects.
Collapse
Affiliation(s)
- David E Gorla
- Universidad Nacional de Córdoba, Instituto de Diversidad y Ecología Animal, CONICET, Córdoba, Argentina
| | - Zhou Xiao-Nong
- Shanghai Jiao Tong University, Chinese Centre for Tropical Diseases Research, National Institute of Parasitic Diseases, One Health Centre, Shanghai, China
| | - Lileia Diotaiuti
- Fundação Oswaldo Cruz-Fiocruz, Instituto René Rachou, Belo Horizonte, MG, Brasil
| | - Pham Thi Khoa
- Science Services of Insect Joint Stock Company, Nam Tu Liem district, Ha Noi, Viet Nam
| | - Etienne Waleckx
- Université de Montpellier, Institut de Recherche pour le Développement, Centre de Coopération Internationale en Recherche Agronomique pour le Développement, Unité Mixte de Recherche, Interactions in the Neglected Tropical Diseases due to Trypanosomatids, Montpellier, France
- Universidad Autónoma de Yucatán, Centro de Investigaciones Regionales Hideyo Noguchi, Mérida, Yucatán, México
| | | | - Liu Qin
- Shanghai Jiao Tong University, Chinese Centre for Tropical Diseases Research, National Institute of Parasitic Diseases, One Health Centre, Shanghai, China
| | - Truong Xuan Lam
- Institute of Ecology and Biological Resources, Vietnam Academy of Science and Technology, Ha Noi, Vietnam
| | - Hector Freilij
- Hospital de Niños Ricardo Gutiérrez, Servicio de Parasitología y Chagas, Buenos Aires, Argentina
| |
Collapse
|
22
|
Muñoz-Calderón AA, Besuschio SA, Wong S, Fernández M, García Cáceres LJ, Giorgio P, Barcan LA, Markham C, Liu YE, de Noya BA, Longhi SA, Schijman AG. Loop-Mediated Isothermal Amplification of Trypanosoma cruzi DNA for Point-of-Care Follow-Up of Anti-Parasitic Treatment of Chagas Disease. Microorganisms 2022; 10:microorganisms10050909. [PMID: 35630354 PMCID: PMC9142941 DOI: 10.3390/microorganisms10050909] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/06/2022] [Accepted: 04/11/2022] [Indexed: 02/05/2023] Open
Abstract
A loop-mediated isothermal amplification assay was evaluated as a surrogate marker of treatment failure in Chagas disease (CD). A convenience series of 18 acute or reactivated CD patients who received anti-parasitic treatment with benznidazole was selected-namely, nine orally infected patients: three people living with HIV and CD reactivation, five chronic CD recipients with reactivation after organ transplantation and one seronegative recipient of a kidney and liver transplant from a CD donor. Fifty-four archival samples (venous blood treated with EDTA or guanidinium hydrochloride-EDTA buffer and cerebrospinal fluid) were extracted using a Spin-column manual kit and tested by T. cruzi Loopamp kit (Tc-LAMP, index test) and standardized real-time PCR (qPCR, comparator test). Of them, 23 samples were also extracted using a novel repurposed 3D printer designed for point-of-care DNA extraction (PrintrLab). The agreement between methods was estimated by Cohen's kappa index and Bland-Altman plot analysis. The T. cruzi Loopamp kit was as sensitive as qPCR for detecting parasite DNA in samples with parasite loads higher than 0.5 parasite equivalents/mL and infected with different discrete typing units. The agreement between qPCR and Tc-LAMP (Spin-column) or Tc-LAMP (PrintrLab) was excellent, with a mean difference of 0.02 [CI = -0.58-0.62] and -0.04 [CI = -0.45-0.37] and a Cohen's kappa coefficient of 0.78 [CI = 0.60-0.96] and 0.90 [CI = 0.71 to 1.00], respectively. These findings encourage prospective field studies to validate the use of LAMP as a surrogate marker of treatment failure in CD.
Collapse
Affiliation(s)
- Arturo A Muñoz-Calderón
- Laboratorio de Biología Molecular de la Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI-CONICET), Buenos Aires 1428, Argentina
| | - Susana A Besuschio
- Laboratorio de Biología Molecular de la Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI-CONICET), Buenos Aires 1428, Argentina
| | - Season Wong
- AI Biosciences, Inc., College Station, TX 77845, USA
| | - Marisa Fernández
- Hospital de Enfermedades Infecciosas "Dr. Francisco J. Muñiz", Buenos Aires 1282, Argentina
| | - Lady J García Cáceres
- Laboratorio de Biología Molecular de la Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI-CONICET), Buenos Aires 1428, Argentina
| | - Patricia Giorgio
- Servicio de Infectología, Hospital Británico de Buenos Aires, Buenos Aires 1280, Argentina
| | - Laura A Barcan
- Sección Infectología, Departamento de Medicina, Hospital Italiano, Buenos Aires 1199, Argentina
| | - Cole Markham
- AI Biosciences, Inc., College Station, TX 77845, USA
| | - Yanwen E Liu
- AI Biosciences, Inc., College Station, TX 77845, USA
| | | | - Silvia A Longhi
- Laboratorio de Biología Molecular de la Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI-CONICET), Buenos Aires 1428, Argentina
| | - Alejandro G Schijman
- Laboratorio de Biología Molecular de la Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI-CONICET), Buenos Aires 1428, Argentina
| |
Collapse
|
23
|
Worldwide Control and Management of Chagas Disease in a New Era of Globalization: a Close Look at Congenital Trypanosoma cruzi Infection. Clin Microbiol Rev 2022; 35:e0015221. [PMID: 35239422 PMCID: PMC9020358 DOI: 10.1128/cmr.00152-21] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Population movements have turned Chagas disease (CD) into a global public health problem. Despite the successful implementation of subregional initiatives to control vectorial and transfusional Trypanosoma cruzi transmission in Latin American settings where the disease is endemic, congenital CD (cCD) remains a significant challenge. In countries where the disease is not endemic, vertical transmission plays a key role in CD expansion and is the main focus of its control. Although several health organizations provide general protocols for cCD control, its management in each geopolitical region depends on local authorities, which has resulted in a multitude of approaches. The aims of this review are to (i) describe the current global situation in CD management, with emphasis on congenital infection, and (ii) summarize the spectrum of available strategies, both official and unofficial, for cCD prevention and control in countries of endemicity and nonendemicity. From an economic point of view, the early detection and treatment of cCD are cost-effective. However, in countries where the disease is not endemic, national health policies for cCD control are nonexistent, and official regional protocols are scarce and restricted to Europe. Countries of endemicity have more protocols in place, but the implementation of diagnostic methods is hampered by economic constraints. Moreover, most protocols in both countries where the disease is endemic and those where it is not endemic have yet to incorporate recently developed technologies. The wide methodological diversity in cCD diagnostic algorithms reflects the lack of a consensus. This review may represent a first step toward the development of a common strategy, which will require the collaboration of health organizations, governments, and experts in the field.
Collapse
|
24
|
Ex Vivo Infection of Human Placental Explants by Trypanosoma cruzi Reveals a microRNA Profile Similar to That Seen in Trophoblast Differentiation. Pathogens 2022; 11:pathogens11030361. [PMID: 35335686 PMCID: PMC8952303 DOI: 10.3390/pathogens11030361] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/09/2022] [Accepted: 03/14/2022] [Indexed: 02/04/2023] Open
Abstract
Congenital Chagas disease, caused by the protozoan parasite Trypanosoma cruzi, is responsible for 22.5% of new cases each year. However, placental transmission occurs in only 5% of infected mothers and it has been proposed that the epithelial turnover of the trophoblast can be considered a local placental defense against the parasite. Thus, Trypanosoma cruzi induces cellular proliferation, differentiation, and apoptotic cell death in the trophoblast, which are regulated, among other mechanisms, by small non-coding RNAs such as microRNAs. On the other hand, ex vivo infection of human placental explants induces a specific microRNA profile that includes microRNAs related to trophoblast differentiation such as miR-512-3p miR-515-5p, codified at the chromosome 19 microRNA cluster. Here we determined the expression validated target genes of miR-512-3p and miR-515-5p, specifically human glial cells missing 1 transcription factor and cellular FLICE-like inhibitory protein, as well as the expression of the main trophoblast differentiation marker human chorionic gonadotrophin during ex vivo infection of human placental explants, and examined how the inhibition or overexpression of both microRNAs affects parasite infection. We conclude that Trypanosoma cruzi-induced trophoblast epithelial turnover, particularly trophoblast differentiation, is at least partially mediated by placenta-specific miR-512-3p and miR-515-5p and that both miRNAs mediate placental susceptibility to ex vivo infection of human placental explants. Knowledge about the role of parasite-modulated microRNAs in the placenta might enable their use as biomarkers, as prognostic and therapeutic tools for congenital Chagas disease in the future.
Collapse
|
25
|
Sousa ASD, Vermeij D, Parra-Henao G, Lesmo V, Fernández EF, Aruni JJC, Mendes FDSNS, Bohorquez LC, Luquetti AO. The CUIDA Chagas Project: towards the elimination of congenital transmission of Chagas disease in Bolivia, Brazil, Colombia, and Paraguay. Rev Soc Bras Med Trop 2022; 55:e01712022. [PMID: 35522804 PMCID: PMC9070074 DOI: 10.1590/0037-8682-0171-2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/08/2022] [Indexed: 11/25/2022] Open
Abstract
Background: Mother-to-child transmission of Chagas disease (CD) has become a relevant problem in both endemic and non-endemic areas. Methods: Description of the CUIDA Chagas Project - Communities United for Innovation, Development and Attention for Chagas disease’. Results: Through innovative and strategic research, this project will provide improved diagnostic and treatment options as well as replicable implementation models that are adaptable to different contexts. Conclusions: By integrating test, treat and care actions for CD into primary health care practices, the burden of CD on people and health systems may be significantly reduced.
Collapse
Affiliation(s)
| | | | | | - Vidalia Lesmo
- Servicio Nacional de Erradicación del Paludismo, Paraguay
| | | | | | | | | | | |
Collapse
|
26
|
Volta BJ, Bustos PL, González C, Natale MA, Perrone AE, Milduberger N, Laucella SA, Bua J. Circulating Cytokine and Chemokine Profiles of Trypanosoma cruzi-Infected Women During Pregnancy and Its Association With Congenital Transmission. J Infect Dis 2021; 224:1086-1095. [PMID: 33528501 DOI: 10.1093/infdis/jiab057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 01/27/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Trypanosoma cruzi, the causative agent of Chagas disease, can be transmitted to the offspring of infected women, which constitutes an epidemiologically significant parasite transmission route in nonendemic areas. It is relevant to evaluate differentially expressed factors in T. cruzi-infected pregnant women as potential markers of Chagas congenital transmission. METHODS Circulating levels of 12 cytokines and chemokines were measured by enzyme-linked immunosorbent assay or cytometric bead array in T. cruzi-infected and uninfected pregnant women in their second trimester of pregnancy and control groups of T. cruzi-infected and uninfected nonpregnant women. RESULTS Trypanosoma cruzi-infected women showed a proinflammatory Th1-biased profile, with increased levels of tumor necrosis factor (TNF)-α, interleukin (IL)-12p70, IL-15, and monokine induced by interferon-gamma (MIG). Uninfected pregnant women presented a biased response towards Th2/Th17/Treg profiles, with increased plasma levels of IL-5, IL-6, IL-1β, IL-17A, and IL-10. Finally, we identified that high parasitemia together with low levels of TNF-α, IL-15, and IL-17, low TNF-α/IL-10 ratio, and high IL-12p70 levels are factors associated with an increased probability of Chagas congenital transmission. CONCLUSIONS Trypanosoma cruzi-infected pregnant women who did not transmit the infection to their babies exhibited a distinct proinflammatory cytokine profile that might serve as a potential predictive marker of congenital transmission.
Collapse
Affiliation(s)
- Bibiana J Volta
- Instituto Nacional de Parasitología Dr. M. Fatala Chaben, Buenos Aires, Argentina.,Administración Nacional de Laboratorios e Institutos de Salud Dr. C.G. Malbrán, Buenos Aires, Argentina
| | - Patricia L Bustos
- Instituto Nacional de Parasitología Dr. M. Fatala Chaben, Buenos Aires, Argentina.,Administración Nacional de Laboratorios e Institutos de Salud Dr. C.G. Malbrán, Buenos Aires, Argentina
| | - Carolina González
- Instituto Nacional de Parasitología Dr. M. Fatala Chaben, Buenos Aires, Argentina.,Administración Nacional de Laboratorios e Institutos de Salud Dr. C.G. Malbrán, Buenos Aires, Argentina.,Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Buenos Aires, Argentina
| | - María Ailén Natale
- Instituto Nacional de Parasitología Dr. M. Fatala Chaben, Buenos Aires, Argentina.,Administración Nacional de Laboratorios e Institutos de Salud Dr. C.G. Malbrán, Buenos Aires, Argentina
| | - Alina E Perrone
- Instituto Nacional de Parasitología Dr. M. Fatala Chaben, Buenos Aires, Argentina.,Administración Nacional de Laboratorios e Institutos de Salud Dr. C.G. Malbrán, Buenos Aires, Argentina
| | - Natalia Milduberger
- Instituto Nacional de Parasitología Dr. M. Fatala Chaben, Buenos Aires, Argentina.,Administración Nacional de Laboratorios e Institutos de Salud Dr. C.G. Malbrán, Buenos Aires, Argentina
| | - Susana A Laucella
- Instituto Nacional de Parasitología Dr. M. Fatala Chaben, Buenos Aires, Argentina.,Administración Nacional de Laboratorios e Institutos de Salud Dr. C.G. Malbrán, Buenos Aires, Argentina
| | - Jacqueline Bua
- Instituto Nacional de Parasitología Dr. M. Fatala Chaben, Buenos Aires, Argentina.,Administración Nacional de Laboratorios e Institutos de Salud Dr. C.G. Malbrán, Buenos Aires, Argentina.,Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Buenos Aires, Argentina
| |
Collapse
|
27
|
Alonso-Padilla J, López MC, Esteva M, Zrein M, Casellas A, Gómez I, Granjon E, Méndez S, Benítez C, Ruiz AM, Sanz S, Gascón J, Thomas MC, Pinazo MJ, Abril M, de Noya BA, Jorge TA, Chatelain E, Grijalva MJ, Guhl F, Hasslocher-Moreno AM, Luquetti AO, Noya O, Ramsey JM, Ribeiro I, Longhi SA, Schijman AG, Sosa-Estani S, Torrico F, Viotti R. Serological reactivity against T. cruzi-derived antigens: Evaluation of their suitability for the assessment of response to treatment in chronic Chagas disease. Acta Trop 2021; 221:105990. [PMID: 34090864 DOI: 10.1016/j.actatropica.2021.105990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 05/20/2021] [Accepted: 05/26/2021] [Indexed: 10/21/2022]
Abstract
Chagas disease, caused by the protozoan Trypanosoma cruzi, affects more than 6 million people worldwide. Following a mostly asymptomatic acute phase, the disease progresses to a long-lasting chronic phase throughout which life-threatening disorders to the heart and/or gastrointestinal tract will manifest in about 30% of those chronically infected. During the chronic phase, the parasitemia is low and intermittent, while a high level of anti-T. cruzi antibodies persist for years. These two features hamper post-chemotherapeutic follow-up of patients with the tools available. The lack of biomarkers for timely assessment of therapeutic response discourages a greater use of the two available anti-parasitic drugs, and complicates the evaluation of new drugs in clinical trials. Herein, we investigated in a blinded case-control study the serological reactivity over time of a group of parasite-derived antigens to potentially address follow up of T. cruzi chronically infected subjects after treatment. We tested PFR2, KMP11, HSP70, 3973, F29 and the InfYnity multiplexed antigenic array, by means of serological assays on a multi-national retrospective collection of samples. Some of the antigens exhibited promising results, underscoring the need for further studies to determine their potential role as treatment response biomarkers.
Collapse
|
28
|
Bisio MMC, Rivero R, Gonzalez N, Ballering G, D'Amico I, Kessler C, Moroni S, Moscatelli G, Ruiz AM, Altcheh J. Diagnostic Accuracy of Two Molecular Tools for Diagnosis of Congenital Chagas Disease. Mol Diagn Ther 2021; 25:791-801. [PMID: 34426953 PMCID: PMC8382099 DOI: 10.1007/s40291-021-00553-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND OBJECTIVE The real prevalence of congenital Chagas disease is undefined because of difficulties in the detection of Trypanosoma cruzi by microscopic examination. The aim of this study was to determine the diagnostic accuracy of two molecular diagnostic tools, qPCR and LAMP, in the diagnosis of congenital Chagas disease in a clinical setting. METHODS To this end, we conducted a prospective cohort study in a tertiary care center, of infants under 9 months of age, born in Buenos Aires to women with Chagas disease. Blood samples were collected for microscopic examination and molecular diagnosis at baseline. If negative, infants were followed up until 9 months of age to determine a final diagnosis by serology. In-house qPCR and LAMP previously validated were challenged as index tests. RESULTS A total of 154 participants were potentially eligible, 120 of whom were enrolled. Finally, 102 (66.2%) of them fulfilled the follow-up. The diagnosis of congenital Chagas disease was confirmed in 13 infants and excluded in 89. Both the sensitivity and specificity of the qPCR were 100.0% (95% confidence interval 75.3-100.0 and 95% confidence interval 95.9-100.0, respectively), whereas the sensitivity and specificity of LAMP were 69.2% (95% confidence interval 38.6-90.9) and 100% (95% confidence interval 95.9-100.0), respectively. CONCLUSIONS The qPCR agreed with the current diagnostic algorithm, and was a reliable and sensitive tool to detect congenital Chagas disease earlier, providing an appropriate and timely identification of infected infants requiring treatment. LAMP was able to detect congenital Chagas disease in infected infants by naked-eye visualization in accordance with a microscopic examination. The advantages of molecular diagnostic tools should be taken into account by the health system to improve congenital Chagas disease diagnosis.
Collapse
Affiliation(s)
- Margarita María Catalina Bisio
- Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños "Dr. Ricardo Gutiérrez", CP 1425 Gallo 1330, Buenos Aires, Argentina. .,Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET-GCBA), Buenos Aires, Argentina.
| | - Rocío Rivero
- Instituto Nacional de Parasitología Dr. Mario Fatala Chaben, Buenos Aires, Argentina
| | - Nicolás Gonzalez
- Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños "Dr. Ricardo Gutiérrez", CP 1425 Gallo 1330, Buenos Aires, Argentina
| | - Griselda Ballering
- Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños "Dr. Ricardo Gutiérrez", CP 1425 Gallo 1330, Buenos Aires, Argentina
| | - Indira D'Amico
- Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños "Dr. Ricardo Gutiérrez", CP 1425 Gallo 1330, Buenos Aires, Argentina
| | - Camila Kessler
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET-GCBA), Buenos Aires, Argentina
| | - Samanta Moroni
- Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños "Dr. Ricardo Gutiérrez", CP 1425 Gallo 1330, Buenos Aires, Argentina
| | - Guillermo Moscatelli
- Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños "Dr. Ricardo Gutiérrez", CP 1425 Gallo 1330, Buenos Aires, Argentina.,Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET-GCBA), Buenos Aires, Argentina
| | - Andrés Mariano Ruiz
- Instituto Nacional de Parasitología Dr. Mario Fatala Chaben, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Jaime Altcheh
- Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños "Dr. Ricardo Gutiérrez", CP 1425 Gallo 1330, Buenos Aires, Argentina. .,Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET-GCBA), Buenos Aires, Argentina.
| |
Collapse
|
29
|
Gamba JC, Roldán C, Prochetto E, Lupi G, Bontempi I, Poncini CV, Vermeulen M, Pérez AR, Marcipar I, Cabrera G. Targeting Myeloid-Derived Suppressor Cells to Enhance a Trans-Sialidase-Based Vaccine Against Trypanosoma cruzi. Front Cell Infect Microbiol 2021; 11:671104. [PMID: 34295832 PMCID: PMC8290872 DOI: 10.3389/fcimb.2021.671104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 06/14/2021] [Indexed: 12/16/2022] Open
Abstract
Trypanosoma cruzi (T. cruzi) is a hemoflagellate protozoan parasite that causes Chagas disease, a neglected tropical disease that affects more than 6 million people around the world, mostly in Latin America. Despite intensive research, there is no vaccine available; therefore, new approaches are needed to further improve vaccine efficacy. It is well established that experimental T. cruzi infection induces a marked immunosuppressed state, which includes notably increases of CD11b+ GR-1+ myeloid-derived suppressor cells (MDSCs) in the spleen, liver and heart of infected mice. We previously showed that a trans-sialidase based vaccine (TSf-ISPA) is able to confer protection against a virulent T. cruzi strain, stimulating the effector immune response and decreasing CD11b+ GR-1+ splenocytes significantly. Here, we show that even in the immunological context elicited by the TSf-ISPA vaccine, the remaining MDSCs are still able to influence several immune populations. Depletion of MDSCs with 5 fluorouracil (5FU) at day 15 post-infection notably reshaped the immune response, as evidenced by flow cytometry of spleen cells obtained from mice after 21 days post-infection. After infection, TSf-ISPA-vaccinated and 5FU-treated mice showed a marked increase of the CD8 response, which included an increased expression of CD107a and CD44 markers in CD8+ cultured splenocytes. In addition, vaccinated and MDSC depleted mice showed an increase in the percentage and number of CD4+ Foxp3+ regulatory T cells (Tregs) as well as in the expression of Foxp3+ in CD4+ splenocytes. Furthermore, depletion of MDSCs also caused changes in the percentage and number of CD11chigh CD8α+ dendritic cells as well as in activation/maturation markers such as CD80, CD40 and MHCII. Thus, the obtained results suggest that MDSCs not only play a role suppressing the effector response during T. cruzi infection, but also strongly modulate the immune response in vaccinated mice, even when the vaccine formulation has a significant protective capacity. Although MDSC depletion at day 15 post-infection did not ameliorated survival or parasitemia levels, depletion of MDSCs during the first week of infection caused a beneficial trend in parasitemia and mice survival of vaccinated mice, supporting the possibility to target MDSCs from different approaches to enhance vaccine efficacy. Finally, since we previously showed that TSf-ISPA immunization causes a slight but significant increase of CD11b+ GR-1+ splenocytes, here we also targeted those cells at the stage of immunization, prior to T. cruzi challenge. Notably, 5FU administration before each dose of TSf-ISPA vaccine was able to significantly ameliorate survival and decrease parasitemia levels of TSf-ISPA-vaccinated and infected mice. Overall, this work supports that targeting MDSCs may be a valuable tool during vaccine design against T. cruzi, and likely for other pathologies that are characterized by the subversion of the immune system.
Collapse
Affiliation(s)
- Juan Cruz Gamba
- Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Carolina Roldán
- Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Estefanía Prochetto
- Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Giuliana Lupi
- Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Iván Bontempi
- Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Carolina Verónica Poncini
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mónica Vermeulen
- Laboratorio de Inmunología Oncológica, Instituto de Medicina Experimental (IMEX-CONICET), Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Ana Rosa Pérez
- IDICER-CONICET and Instituto de Inmunología, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Santa Fe, Argentina
| | - Iván Marcipar
- Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Gabriel Cabrera
- Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| |
Collapse
|
30
|
Benatar AF, Danesi E, Besuschio SA, Bortolotti S, Cafferata ML, Ramirez JC, Albizu CL, Scollo K, Baleani M, Lara L, Agolti G, Seu S, Adamo E, Lucero RH, Irazu L, Rodriguez M, Poeylaut-Palena A, Longhi SA, Esteva M, Althabe F, Rojkin F, Bua J, Sosa-Estani S, Schijman AG. Prospective multicenter evaluation of real time PCR Kit prototype for early diagnosis of congenital Chagas disease. EBioMedicine 2021; 69:103450. [PMID: 34186488 PMCID: PMC8243352 DOI: 10.1016/j.ebiom.2021.103450] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 05/29/2021] [Accepted: 06/04/2021] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Current algorithm for Congenital Chagas Disease (cCD) diagnosis is unsatisfactory due to low sensitivity of the parasitological methods. Moreover, loss to follow-up precludes final serodiagnosis after nine months of life in many cases. A duplex TaqMan qPCR kit for Trypanosoma cruzi DNA amplification was prospectively evaluated in umbilical cord (UCB) and peripheral venous blood (PVB) of infants born to CD mothers at endemic and non-endemic sites of Argentina. METHODS We enrolled and followed-up 370 infants; qPCR was compared to gold-standard cCD diagnosis following studies of diagnostic accuracy guidelines. FINDINGS Fourteen infants (3·78%) had cCD. The qPCR sensitivity and specificity were higher in PVB (72·73%, 99·15% respectively) than in UCB (66·67%, 96·3%). Positive and negative predictive values were 80 and 98·73% and 50 and 98·11% for PVB and UCB, respectively. The Areas under the Curve (AUC) of ROC analysis for qPCR and micromethod (MM) were 0·81 and 0·67 in UCB and 0·86 and 0·68 in PVB, respectively. Parasitic loads ranged from 37·5 to 23,709 parasite equivalents/mL. Discrete typing Unit Tc V was identified in five cCD patients and in six other cCD cases no distinction among Tc II, Tc V or Tc VI was achieved. INTERPRETATION This first prospective field study demonstrated that qPCR was more sensitive than MM for early cCD detection and more accurate in PVB than in UCB. Its use, as an auxiliary diagnostic tool to MM will provide more accurate records on cCD incidence. FUNDING FITS SALUD 001-CHAGAS (FONARSEC, MINCyT, Argentina) to the Public-Private Consortium (INGEBI-CONICET, INP-ANLIS MALBRAN and Wiener Laboratories); ERANET-LAC-HD 328 to AGS and PICT 2015-0074 (FONCYT, MinCyT) to AGS and FA.
Collapse
Affiliation(s)
- Alejandro Francisco Benatar
- Laboratorio de Biología Molecular de la Enfermedad de Chagas (LaBMECh) Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr Héctor Torres” (INGEBI) - Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET; Ciudad Autónoma de Buenos Aires, Argentina
| | - Emmaría Danesi
- Centro Nacional de Diagnóstico e Investigación en Endemoepidemias (ANLIS Dr. C. G. Malbrán, Ciudad Autónoma de Buenos Aires, Argentina)
| | - Susana Alicia Besuschio
- Laboratorio de Biología Molecular de la Enfermedad de Chagas (LaBMECh) Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr Héctor Torres” (INGEBI) - Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET; Ciudad Autónoma de Buenos Aires, Argentina
| | - Santiago Bortolotti
- Centro de Investigación y Biotecnología (CIBIO) de Wiener Laboratorios SAIC, Rosario, Pcia. de Santa Fe, Argentina
| | - María Luisa Cafferata
- Departamento en Salud de la Madre y el Niño, Instituto de Efectividad Clínica y Sanitaria, Ciudad Autónoma de Buenos Aires, Argentina
| | - Juan Carlos Ramirez
- Laboratorio de Biología Molecular de la Enfermedad de Chagas (LaBMECh) Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr Héctor Torres” (INGEBI) - Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET; Ciudad Autónoma de Buenos Aires, Argentina
| | - Constanza Lopez Albizu
- Instituto Nacional de Parasitología Dr. M. Fatala Chabén; Administración Nacional de Laboratorios e Institutos de Salud Carlos G. Malbrán (ANLIS-Malbrán); Ciudad Autónoma de Buenos Aires, Argentina
| | - Karenina Scollo
- Instituto Nacional de Parasitología Dr. M. Fatala Chabén; Administración Nacional de Laboratorios e Institutos de Salud Carlos G. Malbrán (ANLIS-Malbrán); Ciudad Autónoma de Buenos Aires, Argentina
| | - María Baleani
- Centro de Investigación y Biotecnología (CIBIO) de Wiener Laboratorios SAIC, Rosario, Pcia. de Santa Fe, Argentina
| | - Laura Lara
- Instituto de Maternidad y Ginecología "Nuestra Señora de las Mercedes", San Miguel de Tucumán, Pcia. de Tucumán, Argentina
| | - Gustavo Agolti
- Hospital "Dr. Julio César Perrando", Resistencia, Pcia. de Chaco, Argentina
| | - Sandra Seu
- Hospital Regional "Dr. Ramón Carrillo", Santiago del Estero, Pcia. de Santiago del Estero, Argentina
| | - Elsa Adamo
- Centro Integral de Salud La Banda, La Banda, Pcia. de Santiago del Estero, Argentina
| | - Raúl Horacio Lucero
- Instituto de Medicina Regional, Universidad Nacional del Nordeste (IMR-UNNE) Resistencia, Provincia de Chaco, Argentina
| | - Lucía Irazu
- Departamento de Calidad. Administración Nacional de Laboratorios e Institutos de Salud Carlos G. Malbrán (ANLIS-Malbrán)
| | - Marcelo Rodriguez
- Departamento de Calidad. Administración Nacional de Laboratorios e Institutos de Salud Carlos G. Malbrán (ANLIS-Malbrán)
| | - Andrés Poeylaut-Palena
- Centro de Investigación y Biotecnología (CIBIO) de Wiener Laboratorios SAIC, Rosario, Pcia. de Santa Fe, Argentina
| | - Silvia Andrea Longhi
- Laboratorio de Biología Molecular de la Enfermedad de Chagas (LaBMECh) Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr Héctor Torres” (INGEBI) - Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET; Ciudad Autónoma de Buenos Aires, Argentina
| | - Mónica Esteva
- Instituto Nacional de Parasitología Dr. M. Fatala Chabén; Administración Nacional de Laboratorios e Institutos de Salud Carlos G. Malbrán (ANLIS-Malbrán); Ciudad Autónoma de Buenos Aires, Argentina
| | - Fernando Althabe
- Departamento en Salud de la Madre y el Niño, Instituto de Efectividad Clínica y Sanitaria, Ciudad Autónoma de Buenos Aires, Argentina
| | - Federico Rojkin
- Centro de Investigación y Biotecnología (CIBIO) de Wiener Laboratorios SAIC, Rosario, Pcia. de Santa Fe, Argentina
| | - Jacqueline Bua
- Instituto Nacional de Parasitología Dr. M. Fatala Chabén; Administración Nacional de Laboratorios e Institutos de Salud Carlos G. Malbrán (ANLIS-Malbrán); Ciudad Autónoma de Buenos Aires, Argentina
| | - Sergio Sosa-Estani
- Departamento en Salud de la Madre y el Niño, Instituto de Efectividad Clínica y Sanitaria, Ciudad Autónoma de Buenos Aires, Argentina
- Instituto Nacional de Parasitología Dr. M. Fatala Chabén; Administración Nacional de Laboratorios e Institutos de Salud Carlos G. Malbrán (ANLIS-Malbrán); Ciudad Autónoma de Buenos Aires, Argentina
| | - Alejandro Gabriel Schijman
- Laboratorio de Biología Molecular de la Enfermedad de Chagas (LaBMECh) Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr Héctor Torres” (INGEBI) - Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET; Ciudad Autónoma de Buenos Aires, Argentina
| | - Congenital Chagas Disease Study Group
- Laboratorio de Biología Molecular de la Enfermedad de Chagas (LaBMECh) Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr Héctor Torres” (INGEBI) - Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET; Ciudad Autónoma de Buenos Aires, Argentina
- Centro Nacional de Diagnóstico e Investigación en Endemoepidemias (ANLIS Dr. C. G. Malbrán, Ciudad Autónoma de Buenos Aires, Argentina)
- Centro de Investigación y Biotecnología (CIBIO) de Wiener Laboratorios SAIC, Rosario, Pcia. de Santa Fe, Argentina
- Departamento en Salud de la Madre y el Niño, Instituto de Efectividad Clínica y Sanitaria, Ciudad Autónoma de Buenos Aires, Argentina
- Instituto Nacional de Parasitología Dr. M. Fatala Chabén; Administración Nacional de Laboratorios e Institutos de Salud Carlos G. Malbrán (ANLIS-Malbrán); Ciudad Autónoma de Buenos Aires, Argentina
- Instituto de Maternidad y Ginecología "Nuestra Señora de las Mercedes", San Miguel de Tucumán, Pcia. de Tucumán, Argentina
- Hospital "Dr. Julio César Perrando", Resistencia, Pcia. de Chaco, Argentina
- Hospital Regional "Dr. Ramón Carrillo", Santiago del Estero, Pcia. de Santiago del Estero, Argentina
- Centro Integral de Salud La Banda, La Banda, Pcia. de Santiago del Estero, Argentina
- Instituto de Medicina Regional, Universidad Nacional del Nordeste (IMR-UNNE) Resistencia, Provincia de Chaco, Argentina
- Departamento de Calidad. Administración Nacional de Laboratorios e Institutos de Salud Carlos G. Malbrán (ANLIS-Malbrán)
| |
Collapse
|
31
|
Klein MD, Proaño A, Noazin S, Sciaudone M, Gilman RH, Bowman NM. Risk factors for vertical transmission of Chagas disease: A systematic review and meta-analysis. Int J Infect Dis 2021; 105:357-373. [PMID: 33618005 PMCID: PMC8370023 DOI: 10.1016/j.ijid.2021.02.074] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Vertical transmission of Trypanosoma cruzi infection from mother to infant accounts for a growing proportion of new Chagas disease cases. However, no systematic reviews of risk factors for T. cruzi vertical transmission have been performed. METHODS We performed a systematic review of the literature in PubMed, LILACS, and Embase databases, following PRISMA guidelines. Studies were not excluded based on language, country of origin, or publication date. RESULTS Our literature review yielded 27 relevant studies examining a wide variety of risk factors, including maternal age, parasitic load, immunologic factors and vector exposure. Several studies suggested that mothers with higher parasitic loads may have a greater risk of vertical transmission. A meta-analysis of 2 studies found a significantly higher parasitic load among transmitting than non-transmitting mothers with T. cruzi infection. A second meta-analysis of 10 studies demonstrated that maternal age was not significantly associated with vertical transmission risk. CONCLUSIONS The literature suggests that high maternal parasitic load may be a risk factor for congenital Chagas disease among infants of T. cruzi seropositive mothers. Given the considerable heterogeneity and risk of bias among current literature, additional studies are warranted to assess potential risk factors for vertical transmission of T. cruzi infection.
Collapse
Affiliation(s)
- Melissa D Klein
- Department of Medicine, Division of Infectious Diseases, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
| | - Alvaro Proaño
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, USA
| | - Sassan Noazin
- Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Michael Sciaudone
- Department of Medicine, Division of Infectious Diseases, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Robert H Gilman
- Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Natalie M Bowman
- Department of Medicine, Division of Infectious Diseases, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
32
|
Colombo V, Giacomelli A, Casazza G, Galimberti L, Bonazzetti C, Sabaini F, Ridolfo AL, Antinori S. Trypanosoma cruzi infection in Latin American pregnant women living outside endemic countries and frequency of congenital transmission: a systematic review and meta-analysis. J Travel Med 2021; 28:5908540. [PMID: 32946555 DOI: 10.1093/jtm/taaa170] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Chagas disease, as a consequence of globalization and immigration, is no more restricted to Central and Latin America. Therefore, congenital transmission represents a growing public health concern in non-endemic countries. METHODS The aim of this study was to assess the prevalence of Trypanosoma cruzi infection in pregnant Latin American (LA) women living outside endemic countries and the rate of congenital transmission. Data were extracted from studies indexed in PubMed, Scopus, Embase, Lilacs and SciELO databases without language restriction. Two investigators independently collected data on study characteristics, diagnosis, prevalence of infection in pregnant women and congenital infection rate. The data were pooled using a random effects model. RESULTS The search identified 1078 articles of which 29 were eligible regarding prevalence of T. cruzi infection among pregnant women and 1795 articles of which 32 were eligible regarding the congenital transmission rate. The estimated pooled prevalence of T. cruzi infection in LA pregnant women was 4.2% [95% confidence interval (CI): 3.0-5.5]. The prevalence of T. cruzi infection in pregnant women from Bolivia was 15.5% (95% CI: 11.7-19.7) and 0.5% (95% CI: 0.2-0.89) for those coming from all other LA countries. The estimated global rate of congenital transmission was 3.5% (95% CI: 2.5-4.5); excluding poor-quality studies, the rate of congenital transmission was 3.8% (95% CI: 2.4-5.1). CONCLUSIONS Prevalence of Chagas disease among LA pregnant women living outside endemic countries is high, particularly in Bolivian women. The rate of vertical transmission of T. cruzi infection is similar to the rate reported in South and Central American countries.
Collapse
Affiliation(s)
- Valeria Colombo
- Luigi Sacco Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Andrea Giacomelli
- Luigi Sacco Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy.,III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Giovanni Casazza
- Luigi Sacco Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Laura Galimberti
- III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Cecilia Bonazzetti
- Luigi Sacco Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy.,III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Federico Sabaini
- Luigi Sacco Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Anna Lisa Ridolfo
- III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Spinello Antinori
- Luigi Sacco Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy.,III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| |
Collapse
|
33
|
Wehrendt DP, Alonso-Padilla J, Liu B, Rojas Panozo L, Rivera Nina S, Pinto L, Lozano D, Picado A, Abril M, Pinazo MJ, Gascon J, Torrico F, Wong S, Schijman AG. Development and Evaluation of a Three-Dimensional Printer-Based DNA Extraction Method Coupled to Loop Mediated Isothermal Amplification for Point-of-Care Diagnosis of Congenital Chagas Disease in Endemic Regions. J Mol Diagn 2020; 23:389-398. [PMID: 33387697 DOI: 10.1016/j.jmoldx.2020.12.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/22/2020] [Accepted: 12/16/2020] [Indexed: 12/17/2022] Open
Abstract
Vertical transmission of Trypanosomacruzi is the cause of congenital Chagas disease, a re-emerging infectious disease that affects endemic and nonendemic regions alike. An early diagnosis is crucial because prompt treatment achieves a high cure rate, precluding evolution to symptomatic chronic Chagas disease. However, early diagnosis involves low-sensitive parasitologic assays, making necessary serologic confirmation after 9 months of life. With the aim of implementing early diagnostic strategies suitable for minimally equipped laboratories, a T. cruzi-loop-mediated isothermal amplification (LAMP) prototype was coupled with an automated DNA-extraction device repurposed from a three-dimensional printer (PrintrLab). The whole process takes <3 hours to yield a result, with an analytical sensitivity of 0.1 to 2 parasite equivalents per milliliter, depending on the T. cruzi strain. Twenty-five blood samples from neonates born to seropositive mothers were tested blindly. In comparison to quantitative real-time PCR, the PrintrLab-LAMP dual strategy showed high agreement, while both molecular-based methodologies yielded optimal sensitivity and specificity with respect to microscopy-based diagnosis of congenital Chagas disease. PrintrLab-LAMP detected all 10 congenitally transmitted T. cruzi infections, showing promise for point-of-care early diagnosis of congenital Chagas disease.
Collapse
Affiliation(s)
- Diana P Wehrendt
- Laboratorio de Biología Molecular de La Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres", Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Julio Alonso-Padilla
- Barcelona Institute for Global Health, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Bo Liu
- AI Biosciences Inc., College Station, Texas
| | - Lizeth Rojas Panozo
- Fundación Cienciasy Estudios Aplicados para el Desarrollo en Saludy Medio Ambiente, Cochabamba, Bolivia
| | - Silvia Rivera Nina
- Fundación Cienciasy Estudios Aplicados para el Desarrollo en Saludy Medio Ambiente, Cochabamba, Bolivia
| | - Lilian Pinto
- Fundación Cienciasy Estudios Aplicados para el Desarrollo en Saludy Medio Ambiente, Cochabamba, Bolivia
| | - Daniel Lozano
- Fundación Cienciasy Estudios Aplicados para el Desarrollo en Saludy Medio Ambiente, Cochabamba, Bolivia
| | - Albert Picado
- Foundation for Innovative Diagnostics, Geneva, Switzerland
| | | | - Maria J Pinazo
- Barcelona Institute for Global Health, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Joaquim Gascon
- Barcelona Institute for Global Health, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Faustino Torrico
- Fundación Cienciasy Estudios Aplicados para el Desarrollo en Saludy Medio Ambiente, Cochabamba, Bolivia
| | - Season Wong
- AI Biosciences Inc., College Station, Texas.
| | - Alejandro G Schijman
- Laboratorio de Biología Molecular de La Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres", Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
34
|
Klein MD, Tinajeros F, Del Carmen Menduiña M, Málaga E, Condori BJ, Verástegui M, Urquizu F, Gilman RH, Bowman NM. Risk Factors for Maternal Chagas Disease and Vertical Transmission in a Bolivian Hospital. Clin Infect Dis 2020; 73:e2450-e2456. [PMID: 33367656 DOI: 10.1093/cid/ciaa1885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Vertical transmission of Trypanosoma cruzi infection accounts for a growing proportion of new cases of Chagas disease. Better risk stratification is needed to predict which women are more likely to transmit the infection. METHODS This study enrolled women and their infants at the Percy Boland Women's Hospital in Santa Cruz, Bolivia. Pregnant women were screened for Chagas disease by rapid test and received confirmatory serology. Infants of seropositive mothers underwent diagnostic testing with quantitative polymerase chain reaction (qPCR). RESULTS Among 5,828 enrolled women, 1,271 (21.8%) screened positive for Chagas disease. Older maternal age, family history of Chagas disease, home conditions, lower education level, and history of living in a rural area were significantly associated with higher adjusted odds of maternal infection. Of the 1,325 infants of seropositive mothers, 65 infants (4.9%) were diagnosed with congenital Chagas disease. Protective factors against transmission included Cesarean delivery (adjusted OR [aOR]: 0.60, 95% CI: 0.36-0.99) and family history of Chagas disease (aOR: 0.58, 95% CI: 0.34-0.99). Twins were significantly more likely to be congenitally infected than singleton births (OR: 3.32, 95% CI: 1.60-6.90). Among congenitally infected infants, 32.3% had low birth weight, and 30.8% required hospitalization after birth. CONCLUSIONS Although improved access to screening and qPCR increased the number of infants diagnosed with congenital Chagas disease, many infants remain undiagnosed. A better understanding of risk factors and improved access to highly sensitive and specific diagnostic techniques for congenital Chagas disease may help improve regional initiatives to reduce disease burden.
Collapse
Affiliation(s)
- Melissa D Klein
- Department of Medicine, Division of Infectious Diseases, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
| | | | | | - Edith Málaga
- Infectious Diseases Research Laboratory, Department of Cellular and Molecular Sciences, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Beth J Condori
- Infectious Diseases Research Laboratory, Department of Cellular and Molecular Sciences, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Manuela Verástegui
- Infectious Diseases Research Laboratory, Department of Cellular and Molecular Sciences, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Federico Urquizu
- Hospital Percy Boland Rodríguez, Ministerio de Salud Bolivia, Santa Cruz, Bolivia
| | - Robert H Gilman
- Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Natalie M Bowman
- Department of Medicine, Division of Infectious Diseases, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
35
|
de Souza ALADAG, Mesquita CT. Chagas Disease - Past and Future. INTERNATIONAL JOURNAL OF CARDIOVASCULAR SCIENCES 2020. [DOI: 10.36660/ijcs.20200351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
36
|
|
37
|
Besuschio SA, Picado A, Muñoz-Calderón A, Wehrendt DP, Fernández M, Benatar A, Diaz-Bello Z, Irurtia C, Cruz I, Ndung’u JM, Cafferata ML, Montenegro G, Sosa Estani S, Lucero RH, Alarcón de Noya B, Longhi SA, Schijman AG. Trypanosoma cruzi loop-mediated isothermal amplification (Trypanosoma cruzi Loopamp) kit for detection of congenital, acute and Chagas disease reactivation. PLoS Negl Trop Dis 2020; 14:e0008402. [PMID: 32797041 PMCID: PMC7458301 DOI: 10.1371/journal.pntd.0008402] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 08/31/2020] [Accepted: 05/20/2020] [Indexed: 12/15/2022] Open
Abstract
A Trypanosoma cruzi Loopamp kit was recently developed as a ready-to-use diagnostic method requiring minimal laboratory facilities. We evaluated its diagnostic accuracy for detection of acute Chagas disease (CD) in different epidemiological and clinical scenarios. In this retrospective study, a convenience series of clinical samples (venous blood treated with EDTA or different stabilizer agents, heel-prick blood in filter paper or cerebrospinal fluid samples (CSF)) from 30 infants born to seropositive mothers (13 with congenital CD and 17 noninfected), four recipients of organs from CD donors, six orally-infected cases after consumption of contaminated guava juice and six CD patients coinfected with HIV at risk of CD reactivation (N = 46 patients, 46 blood samples and 1 CSF sample) were tested by T. cruzi Loopamp kit (Tc LAMP) and standardized quantitative real-time PCR (qPCR). T. cruzi Loopamp accuracy was estimated using the case definition in the different groups as a reference. Cohen's kappa coefficient (κ) was applied to measure the agreement between Tc LAMP (index test) and qPCR (reference test). Sensitivity and specificity of T. cruzi Loopamp kit in blood samples from the pooled clinical groups was 93% (95% CI: 77-99) and 100% (95% CI: 80-100) respectively. The agreement between Tc LAMP and qPCR was almost perfect (κ = 0.92, 95% CI: 0.62-1.00). The T. cruzi Loopamp kit was sensitive and specific for detection of T. cruzi infection. It was carried out from DNA extracted from peripheral blood samples (via frozen EDTA blood, guanidine hydrochloride-EDTA blood, DNAgard blood and dried blood spots), as well as in CSF specimens infected with TcI or TcII/V/VI parasite populations. The T. cruzi Loopamp kit appears potentially useful for rapid detection of T. cruzi infection in congenital, acute and CD reactivation due to HIV infection.
Collapse
Affiliation(s)
- Susana A. Besuschio
- Laboratorio de Biología Molecular de la Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr Héctor Torres”, (INGEBI-CONICET), Buenos Aires, Argentina
| | - Albert Picado
- Foundation for Innovative New Diagnostics (FIND), Geneva, Switzerland
| | - Arturo Muñoz-Calderón
- Laboratorio de Biología Molecular de la Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr Héctor Torres”, (INGEBI-CONICET), Buenos Aires, Argentina
| | - Diana P Wehrendt
- Laboratorio de Biología Molecular de la Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr Héctor Torres”, (INGEBI-CONICET), Buenos Aires, Argentina
| | - Marisa Fernández
- Hospital de Enfermedades Infecciosas “Dr. Francisco J. Muñiz” Buenos Aires, Argentina
- Instituto Nacional de Parasitología, “Dr Mario Fatala Chabén”, ANLIS CG Malbrán, Buenos Aires, Argentina
| | - Alejandro Benatar
- Laboratorio de Biología Molecular de la Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr Héctor Torres”, (INGEBI-CONICET), Buenos Aires, Argentina
| | - Zoraida Diaz-Bello
- Instituto de Medicina Tropical, Universidad Central de Venezuela, Caracas, Venezuela 5
| | - Cecilia Irurtia
- Hospital Nacional “Profesor Alejandro Posadas”, Villa Sarmiento, Buenos Aires, Argentina
| | - Israel Cruz
- Foundation for Innovative New Diagnostics (FIND), Geneva, Switzerland
- National School of Public Health, Instituto de Salud Carlos III, Madrid, Spain
| | - Joseph M Ndung’u
- Foundation for Innovative New Diagnostics (FIND), Geneva, Switzerland
| | - María L Cafferata
- Departamento en Salud de la Madre y el Niño, Instituto de Efectividad Clínica y Sanitaria – Centro de Investigación en Epidemiología y Salud Pública (IECS-CIESP), Buenos Aires, Argentina
| | - Graciela Montenegro
- Hospital Nacional “Profesor Alejandro Posadas”, Villa Sarmiento, Buenos Aires, Argentina
| | - Sergio Sosa Estani
- Instituto Nacional de Parasitología, “Dr Mario Fatala Chabén”, ANLIS CG Malbrán, Buenos Aires, Argentina
| | - Raúl H. Lucero
- Área de Biología Molecular, Instituto de Medicina Regional, Universidad Nacional del Nordeste, Resistencia, Argentina
| | | | - Silvia A Longhi
- Laboratorio de Biología Molecular de la Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr Héctor Torres”, (INGEBI-CONICET), Buenos Aires, Argentina
| | - Alejandro G Schijman
- Laboratorio de Biología Molecular de la Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr Héctor Torres”, (INGEBI-CONICET), Buenos Aires, Argentina
| |
Collapse
|
38
|
Crudo F, Piorno P, Krupitzki H, Guilera A, López-Albizu C, Danesi E, Scollo K, Lloveras S, Mir S, Álvarez M, Yudis S, Cayo Fernández MA, Cipri D, Krolewiecki A, Pereiro AC, Periago MV, Abril MC, Fernandez M. How to implement the framework for the elimination of mother-to-child transmission of HIV, syphilis, hepatitis B and Chagas (EMTCT Plus) in a disperse rural population from the Gran Chaco region: A tailor-made program focused on pregnant women. PLoS Negl Trop Dis 2020; 14:e0008078. [PMID: 32463835 PMCID: PMC7255590 DOI: 10.1371/journal.pntd.0008078] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Favio Crudo
- Fundación Mundo Sano, Buenos Aires, Argentina
- Asociación para el Desarrollo Sanitario Regional (ADESAR), San Antonio de Areco, Buenos Aires, Argentina
| | - Pablo Piorno
- Asociación para el Desarrollo Sanitario Regional (ADESAR), San Antonio de Areco, Buenos Aires, Argentina
| | - Hugo Krupitzki
- Asociación para el Desarrollo Sanitario Regional (ADESAR), San Antonio de Areco, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Educación Médica e Investigaciones Clínicas "Norberto Quirno" (CEMIC), Buenos Aires, Argentina
| | - Analia Guilera
- Asociación para el Desarrollo Sanitario Regional (ADESAR), San Antonio de Areco, Buenos Aires, Argentina
| | - Constanza López-Albizu
- Asociación para el Desarrollo Sanitario Regional (ADESAR), San Antonio de Areco, Buenos Aires, Argentina
- Instituto de Parasitología "Dr. Mario Fatala Chaben," Administración Nacional de Laboratorios e Institutos de Salud (ANLIS), Buenos Aires, Argentina
| | - Emmaria Danesi
- Asociación para el Desarrollo Sanitario Regional (ADESAR), San Antonio de Areco, Buenos Aires, Argentina
- Centro Nacional de Diagnóstico e Investigaciones en Endemo-epidemias (CeNDIE), ANLIS, Buenos Aires, Argentina
| | - Karerina Scollo
- Instituto de Parasitología "Dr. Mario Fatala Chaben," Administración Nacional de Laboratorios e Institutos de Salud (ANLIS), Buenos Aires, Argentina
| | - Susana Lloveras
- Asociación para el Desarrollo Sanitario Regional (ADESAR), San Antonio de Areco, Buenos Aires, Argentina
| | - Sebastián Mir
- Universidad Nacional de San Antonio de Areco, San Antonio de Areco, Buenos Aires, Argentina
| | | | - Silvio Yudis
- Ministerio de Primera Infancia, Salta, Argentina
| | | | - Diego Cipri
- XVI Región Sanitaria Boquerón, Ministerio de Salud Pública y Bienestar Social, Mariscal José Félix Estigarribia, Paraguay
| | - Alejandro Krolewiecki
- Fundación Mundo Sano, Buenos Aires, Argentina
- Asociación para el Desarrollo Sanitario Regional (ADESAR), San Antonio de Areco, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científica y Técnicas (CONICET), Instituto de Investigaciones en Enfermedades Tropicales (IIET), Universidad de Salta - Sede Regional Orán, San Ramón de Orán, Salta, Argentina
| | | | - María Victoria Periago
- Consejo Nacional de Investigaciones Científica y Técnicas (CONICET), Fundación Mundo Sano, Buenos Aires, Argentina
| | | | - Mariana Fernandez
- Asociación para el Desarrollo Sanitario Regional (ADESAR), San Antonio de Areco, Buenos Aires, Argentina
| |
Collapse
|
39
|
Echeverría LE, Marcus R, Novick G, Sosa-Estani S, Ralston K, Zaidel EJ, Forsyth C, RIbeiro ALP, Mendoza I, Falconi ML, Mitelman J, Morillo CA, Pereiro AC, Pinazo MJ, Salvatella R, Martinez F, Perel P, Liprandi ÁS, Piñeiro DJ, Molina GR. WHF IASC Roadmap on Chagas Disease. Glob Heart 2020; 15:26. [PMID: 32489799 PMCID: PMC7218776 DOI: 10.5334/gh.484] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 01/17/2020] [Indexed: 02/06/2023] Open
Abstract
Background Chagas Disease is a neglected tropical disease caused by the protozoan Trypanosoma cruzi, with some of the most serious manifestations affecting the cardiovascular system. It is a chronic, stigmatizing condition, closely associated with poverty and affecting close to 6 million people globally. Although historically the disease was limited to endemic areas of Latin America recent years have seen an increasing global spread. In addition to the morbidity and mortality associated with the disease, the social and economic burdens on individuals and society are substantial. Often called the 'silent killer', Chagas disease is characterized by a long, asymptomatic phase in affected individuals. Approximately 30% then go on develop chronic Chagas cardiomyopathy and other serious cardiac complications such as stroke, rhythm disturbances and severe heart failure. Methods In a collaboration of the World Hearth Federation (WHF) and the Inter-American Society of Cardiology (IASC) a writing group consisting of 20 diverse experts on Chagas disease (CD) was convened. The group provided up to date expert knowledge based on their area of expertise. An extensive review of the literature describing obstacles to diagnosis and treatment of CD along with proposed solutions was conducted. A survey was sent to all WHF Members and, using snowball sampling to widen the consultation, to a variety of health care professionals working in the CD global health community. The results were analyzed, open comments were reviewed and consolidated, and the findings were incorporated into this document, thus ensuring a consensus representation. Results The WHF IASC Roadmap on Chagas Disease offers a comprehensive summary of current knowledge on prevention, diagnosis and management of the disease. In providing an analysis of 'roadblocks' in access to comprehensive care for Chagas disease patients, the document serves as a framework from which strategies for implementation such as national plans can be formulated. Several dimensions are considered in the analysis: healthcare system capabilities, governance, financing, community awareness and advocacy. Conclusion The WHF IASC Roadmap proposes strategies and evidence-based solutions for healthcare professionals, health authorities and governments to help overcome the barriers to comprehensive care for Chagas disease patients. This roadmap describes an ideal patient care pathway, and explores the roadblocks along the way, offering potential solutions based on available research and examples in practice. It represents a call to action to decision-makers and health care professionals to step up efforts to eradicate Chagas disease.
Collapse
Affiliation(s)
| | - Rachel Marcus
- LASOCHA, Washington DC, US
- Medstar Union Memorial Hospital, Baltimore, MD, US
| | - Gabriel Novick
- Swiss Medical Group, Buenos Aires, AR
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, US
| | - Sergio Sosa-Estani
- Drugs for Neglected Diseases initiative-Latin America, Rio de Janeiro, BR
| | | | - Ezequiel Jose Zaidel
- Sanatorio Güemes, Buenos Aires, AR
- Pharmacology Department, School of Medicine, University of Buenos Aires, Buenos Aires, AR
| | - Colin Forsyth
- Drugs for Neglected Diseases initiative-Latin America, Rio de Janeiro, BR
| | - Antonio Luiz P. RIbeiro
- Internal Medicine Department, School of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, BR
- Hospital das Clínicas, UFMG, Belo Horizonte, BR
| | | | - Mariano Luis Falconi
- Cardiology Division, Italian Hospital of Buenos Aires, Buenos Aires, AR
- University Institute of the Italian Hospital of Buenos Aires, Buenos Aires, AR
| | - Jorge Mitelman
- Faculty of Medicine, University of Buenos Aires, Buenos Aires, AR
- School of Medicine, Barcélo University, Buenos Aires, AR
| | - Carlos A. Morillo
- Department of Cardiac Sciences, Cumming School of Medicine Division of Cardiology, Libin Cardiovascular Institute, University of Calgary, Calgary, CA
- Southeastern Alberta Region, Alberta Health Services, Foothills Medical Centre, CA
| | | | | | | | - Felipe Martinez
- National University of Cordoba, Cordoba, AR
- DAMIC Institute/Rusculleda Foundation, Cordoba, AR
| | - Pablo Perel
- World Heart Federation, Geneva, CH
- Centre for Global Chronic Conditions, London School of Hygiene and Tropical Medicine, London, GB
| | - Álvaro Sosa Liprandi
- Sanatorio Güemes, Buenos Aires, AR
- Medical School of Cardiology, University of Buenos Aires, Buenos Aires, AR
| | | | | |
Collapse
|
40
|
Mann AE, Mitchell EA, Zhang Y, Curtis-Robles R, Thapa S, Hamer SA, Allen MS. Comparison of the Bacterial Gut Microbiome of North American Triatoma spp. With and Without Trypanosoma cruzi. Front Microbiol 2020; 11:364. [PMID: 32231645 PMCID: PMC7082358 DOI: 10.3389/fmicb.2020.00364] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 02/18/2020] [Indexed: 12/20/2022] Open
Abstract
Chagas disease, caused by the hemoflagellate protist Trypanosoma cruzi, affects nearly 6 million people worldwide, mainly in Latin America. Hematophagous triatomine insects (“kissing bugs”) are the primary vectors of T. cruzi throughout the Americas and feed on a variety of animals, including humans. Control of triatomines is central to the control of T. cruzi infection. Recent advances in mitigation of other insect-borne diseases via the manipulation of insect-associated bacteria as a way to halt or slow disease transmission has opened questions to the applicability of these methods to Chagas disease vectors. Few studies have examined the hindgut microbiome of triatomines found in North America. In the current study, two species of triatomines were collected across Texas, United States, screened for the presence of T. cruzi, and analyzed for the bacterial composition of their hindguts using a 16S rRNA gene-fragment metabarcoding approach. We compared diversity of microbial community profiles across 74 triatomine insects to address the hypothesis that the richness and abundance of bacterial groups differ by T. cruzi infection and strain type, blood meal engorgement status, insect species, sex, and collection location. The gut microbial community of individual triatomines was characterized by low intraindividual taxonomic diversity and high interindividual variation that was weakly predicted by triatomine species, and was not predicted by triatomine sex, collection location, T. cruzi infection status, or blood meal score. However, we did find bacterial groups enriched in T. cruzi-positive individuals, including Enterobacterales, and Petrimonas. Additionally, we detected Salmonella enterica subspecies diarizonae in three triatomine individuals; this species is commonly associated with reptiles and domesticated animals and is a pathogen of humans. These data suggest that Triatoma spp. in Texas have variable patterns of colonized and transient bacteria, and may aid in development of novel means to interfere with transmission of the Chagas disease parasite T. cruzi. Deeper understanding of the effects of parasite infection on diverse insect vector microbiomes may highlight disease transmission risk and facilitate discovery of possible intervention strategies for biological control of this emerging vector-borne disease of global health significance.
Collapse
Affiliation(s)
- Allison E Mann
- Tick-Borne Disease Research Laboratory, Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Elizabeth A Mitchell
- Tick-Borne Disease Research Laboratory, Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Yan Zhang
- Tick-Borne Disease Research Laboratory, Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Rachel Curtis-Robles
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, United States
| | - Santosh Thapa
- Tick-Borne Disease Research Laboratory, Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX, United States.,Texas Children's Microbiome Center, Texas Children's Hospital, Houston, TX, United States.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Sarah A Hamer
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, United States
| | - Michael S Allen
- Tick-Borne Disease Research Laboratory, Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX, United States
| |
Collapse
|
41
|
Parra-Henao G, Oliveros H, Hotez PJ, Motoa G, Franco-Paredes C, Henao-Martínez AF. In Search of Congenital Chagas Disease in the Sierra Nevada de Santa Marta, Colombia. Am J Trop Med Hyg 2020; 101:482-483. [PMID: 31264558 DOI: 10.4269/ajtmh.19-0110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Chagas disease remains a major impediment to sustainable socioeconomic development in Latin America. Transplacental transmission explains the persistence of transmission in urban areas, in non-endemic regions, and in areas with an established interrupted vectorial transmission. One of every five cases of congenital Chagas disease in the world occurs in Colombia and Venezuela. The massive migration of impoverished populations from neighboring Venezuela has worsened the situation creating a humanitarian crisis in Northeastern Colombia, including the Sierra Nevada de Santa Marta. The prevalence of Chagas infection among pregnant women in these areas is higher than the national average, and the public health resources are insufficient. This perspective discusses the associated increased morbidity and mortality of congenital Chagas in this region, where stigmatization contributes to the impression among health authorities and the general population that it affects indigenous communities only. The monitoring and control of congenital Chagas disease in the Sierra Nevada of Santa Marta is a public health necessity that demands urgent and effective interventions.
Collapse
Affiliation(s)
- Gabriel Parra-Henao
- Centro de Investigación en Salud para el Trópico (CIST), Universidad Cooperativa de Colombia, Santa Marta, Colombia
| | - Horacio Oliveros
- Centro de Investigación en Salud para el Trópico (CIST), Universidad Cooperativa de Colombia, Santa Marta, Colombia
| | - Peter J Hotez
- Department of Biology, Baylor University, Waco, Texas.,Department of Pediatrics, Texas Children's Hospital Center for Vaccine Development, National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas
| | - Gabriel Motoa
- Division of Infectious Diseases, University of Colorado Denver, School of Medicine, Aurora, Colorado
| | - Carlos Franco-Paredes
- Hospital Infantil de México, Federico Gómez, México City, México.,Division of Infectious Diseases, University of Colorado Denver, School of Medicine, Aurora, Colorado
| | - Andrés F Henao-Martínez
- Division of Infectious Diseases, University of Colorado Denver, School of Medicine, Aurora, Colorado
| |
Collapse
|
42
|
Rios L, Campos EE, Menon R, Zago MP, Garg NJ. Epidemiology and pathogenesis of maternal-fetal transmission of Trypanosoma cruzi and a case for vaccine development against congenital Chagas disease. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165591. [PMID: 31678160 PMCID: PMC6954953 DOI: 10.1016/j.bbadis.2019.165591] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/12/2019] [Accepted: 09/25/2019] [Indexed: 12/11/2022]
Abstract
Trypanos o ma cruzi (T. cruzi or Tc) is the causative agent of Chagas disease (CD). It is common for patients to suffer from non-specific symptoms or be clinically asymptomatic with acute and chronic conditions acquired through various routes of transmission. The expecting women and their fetuses are vulnerable to congenital transmission of Tc. Pregnant women face formidable health challenges because the frontline antiparasitic drugs, benznidazole and nifurtimox, are contraindicated during pregnancy. However, it is worthwhile to highlight that newborns can be cured if they are diagnosed and given treatment in a timely manner. In this review, we discuss the pathogenesis of maternal-fetal transmission of Tc and provide a justification for the investment in the development of vaccines against congenital CD.
Collapse
Affiliation(s)
- Lizette Rios
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - E Emanuel Campos
- Instituto de Patología Experimental, Universidad Nacional de Salta - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Salta, Argentina
| | - Ramkumar Menon
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, USA
| | - M Paola Zago
- Instituto de Patología Experimental, Universidad Nacional de Salta - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Salta, Argentina.
| | - Nisha J Garg
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
43
|
Norman FF, Comeche B, Chamorro S, López-Vélez R. Overcoming challenges in the diagnosis and treatment of parasitic infectious diseases in migrants. Expert Rev Anti Infect Ther 2020; 18:127-143. [PMID: 31914335 DOI: 10.1080/14787210.2020.1713099] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Introduction: Recent increases in population movements have created novel health challenges in many areas of the World, and health policies have been adapted accordingly in several countries. However, screening guidelines for infectious diseases are not standardized and generally do not include comprehensive screening for parasitic infections.Areas covered: Malaria, Chagas disease, leishmaniasis, amebiasis, filariases, strongyloidiasis, and schistosomiasis are reviewed, focusing on the challenges posed for their diagnosis and management in vulnerable populations such as migrants. The methodology included literature searches in public databases such as PubMed.gov and Google Scholar and search of the US National Library of Medicine online database of privately and publicly funded clinical studies (ClinicalTrials.gov) until November 2019.Expert opinion: Parasitic infections which may remain asymptomatic for prolonged periods, leading to chronic infection and complications, and/or may be transmitted in non-endemic areas are ideal candidates for screening. Proposed strategies to improve diagnosis in vulnerable groups such as migrants include facilitating access to healthcare in a multi-dimensional manner considering location, individual characteristics, and timing. Limitations and availability of specific diagnostic techniques should be addressed and focus on drug and vaccine development for these neglected infections should be prioritized through collaborative initiatives with public disclosure of results.
Collapse
Affiliation(s)
- Francesca F Norman
- National Referral Unit for Tropical Diseases, Infectious Diseases Department, Ramón y Cajal University Hospital, IRYCIS, Madrid, Spain
| | - Belen Comeche
- National Referral Unit for Tropical Diseases, Infectious Diseases Department, Ramón y Cajal University Hospital, IRYCIS, Madrid, Spain
| | - Sandra Chamorro
- National Referral Unit for Tropical Diseases, Infectious Diseases Department, Ramón y Cajal University Hospital, IRYCIS, Madrid, Spain
| | - Rogelio López-Vélez
- National Referral Unit for Tropical Diseases, Infectious Diseases Department, Ramón y Cajal University Hospital, IRYCIS, Madrid, Spain
| |
Collapse
|
44
|
Alonso-Vega C, Losada-Galván I, Pinazo MJ, Sancho Mas J, Brustenga JG, Alonso-Padilla J. The senseless orphanage of Chagas disease. Expert Opin Orphan Drugs 2019. [DOI: 10.1080/21678707.2019.1701432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
| | | | | | - Javier Sancho Mas
- ISGlobal - Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | | | | |
Collapse
|
45
|
Simón M, Gil-Gallardo LJ, Asunción Iborra M, Carrilero B, López MC, Romay-Barja M, Murcia L, Carmen Thomas M, Benito A, Segovia M. An observational longitudinal study to evaluate tools and strategies available for the diagnosis of Congenital Chagas Disease in a non-endemic country. Acta Trop 2019; 199:105127. [PMID: 31394076 DOI: 10.1016/j.actatropica.2019.105127] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 07/19/2019] [Accepted: 08/04/2019] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Congenital Chagas Disease (CCD) has become a global health problem. Early diagnosis and treatment is essential for the cure of the disease. Our aim was to evaluate techniques and samples used for the diagnosis of CCD in order to improve diagnostic strategies. METHODS A total of 181 children born in Spain from Latin American Chagas-infected mothers were consecutively enrolled and studied by microhematocrit, PCR and serology tests at 0-2, 6 and 9-12 months of age and followed up when it was required. Samples of cord blood and peripheral blood were collected for T. cruzi detection by PCR. Parasite culture was performed in patients with a positive PCR. RESULTS Of 181 children, 7 children (3.9%) were lost to follow-up. A total of 174 children completed follow-up, 12 were diagnosed with CCD (6.9%) and 162 (93.1%) as uninfected children (negative serology tests at the end of the follow-up). Traditional parasitological diagnosis by microhematocrit had a poor performance (sensitivity was 10%), while PCR in peripheral blood showed high sensitivity (90.9%) and specificity (100%), allowing the early diagnosis of 9 infected children during the first 6-months-old. In the other 3 congenital cases, diagnosis was only possible at 12 months by serological and molecular techniques. However, PCR in cord blood showed low sensitivity (33.3%) and less specificity (96.4%) for the diagnosis. CONCLUSION PCR in peripheral blood has proven to be the most adequate strategy for the diagnosis of CCD, allowing an early and reliable diagnosis.
Collapse
|
46
|
Carlier Y, Altcheh J, Angheben A, Freilij H, Luquetti AO, Schijman AG, Segovia M, Wagner N, Albajar Vinas P. Congenital Chagas disease: Updated recommendations for prevention, diagnosis, treatment, and follow-up of newborns and siblings, girls, women of childbearing age, and pregnant women. PLoS Negl Trop Dis 2019; 13:e0007694. [PMID: 31647811 PMCID: PMC6812740 DOI: 10.1371/journal.pntd.0007694] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Yves Carlier
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, United States of America
- * E-mail:
| | - Jaime Altcheh
- Servicio de Parasitología, Hospital de Niños Ricardo Gutiérrez (Centro colaborador en Chagas pediátrico OPS/OMS), Instituto Multidisciplinario de Investigación en Patologías Pediátricas (IMIPP), CONICET-GCBA, Buenos Aires, Argentina
| | - Andrea Angheben
- Centro per le Malattie Tropicali, IRCCS Ospedale “Sacro Cuore—Don Calabria,” Negrar (Verona), Italy
| | - Hector Freilij
- Servicio de Parasitología, Hospital de Niños Ricardo Gutiérrez (Centro colaborador en Chagas pediátrico OPS/OMS), Instituto Multidisciplinario de Investigación en Patologías Pediátricas (IMIPP), CONICET-GCBA, Buenos Aires, Argentina
| | - Alejandro O. Luquetti
- Laboratório de Chagas, Hospital das Clínicas, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Alejandro G. Schijman
- Laboratorio de Biología Molecular de la Enfermedad de Chagas, INGEBI-CONICET, Buenos Aires, Argentina
| | - Manuel Segovia
- Unidad Regional de Medicina Tropical, Hospital Clínico Universitario Virgen de la Arrixaca, El Palmar (Murcia), Spain
| | - Noemie Wagner
- Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - Pedro Albajar Vinas
- Department of Control of Neglected Tropical Diseases, World Health Organization, Geneva, Switzerland
| |
Collapse
|
47
|
Norman FF, López-Vélez R. Chagas disease: comments on the 2018 PAHO Guidelines for diagnosis and management. J Travel Med 2019; 26:5549353. [PMID: 31407784 DOI: 10.1093/jtm/taz060] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/24/2019] [Accepted: 08/01/2019] [Indexed: 01/12/2023]
Abstract
BACKGROUND Chagas disease, or American trypanosomiasis, is a protozoan infectious disease endemic throughout most of the Americas, caused by the trypanosome, Trypanosoma cruzi, and mainly transmitted to humans by reduviid or kissing bugs. Some progress has been achieved in control of the disease mainly in endemic areas, but migration flows have acted as drivers for the emergence of the disease mainly in non-endemic areas of Europe and North America. Most imported cases of Chagas disease in Europe are reported in migrants from highly endemic areas of countries such as Bolivia and Paraguay, and reports of Chagas disease in travellers are extremely rare. METHODS Pan American Health Organization (PAHO) recently updated their guidelines on the diagnosis and management of Chagas. These guidelines and their applicability to migrants and travellers are reviewed. RESULTS PAHO recommends the use of two serological tests for diagnosis of chronic infection (allowing for the use of a single sensitive test followed by confirmation in special settings such as the screening of potential blood donors). The indication for specific trypanocidal treatment of acute infections, children and women of child-bearing age remains as before, with either benznidazole or nifurtimox being the treatment of choice. For chronic infection with/without organ damage, treatment recommendations are less well defined. Although treatment is generally not recommended in patients with visceral involvement, decisions regarding treatment need to be tailored to the individual. Either benznidazole or nifurtimox may be used for initial treatment. CONCLUSIONS The recent PAHO Guidelines provide a framework to aid the diagnosis and management of this infection, but several aspects such as the underdiagnosis of infections, the multidisciplinary approach to patient management, the investigation of novel biomarkers of disease progression/response to treatment and the development of new treatment strategies are areas which should be further strengthened.
Collapse
Affiliation(s)
- Francesca F Norman
- National Referral Unit for Tropical Diseases, Infectious Diseases Department, Ramón y Cajal University Hospital, IRYCIS, Ctra Colmenar, Km 9,100, 28034, Madrid, Spain
| | - Rogelio López-Vélez
- National Referral Unit for Tropical Diseases, Infectious Diseases Department, Ramón y Cajal University Hospital, IRYCIS, Ctra Colmenar, Km 9,100, 28034, Madrid, Spain
| |
Collapse
|
48
|
Kemmerling U, Osuna A, Schijman AG, Truyens C. Congenital Transmission of Trypanosoma cruzi: A Review About the Interactions Between the Parasite, the Placenta, the Maternal and the Fetal/Neonatal Immune Responses. Front Microbiol 2019; 10:1854. [PMID: 31474955 PMCID: PMC6702454 DOI: 10.3389/fmicb.2019.01854] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 07/29/2019] [Indexed: 12/11/2022] Open
Abstract
Chagas disease (CD), caused by the protozoan parasite Trypanosoma cruzi, is considered a neglected tropical disease by the World Health Organization. Congenital transmission of CD is an increasingly relevant public health problem. It progressively becomes the main transmission route over others and can occur in both endemic and non-endemic countries. Though most congenitally infected newborns are asymptomatic at birth, they display higher frequencies of prematurity, low birth weight, and lower Apgar scores compared to uninfected ones, and some suffer from severe symptoms. If not diagnosed and treated, infected newborns are at risk of developing disabling and life-threatening chronic pathologies later in life. The success or failure of congenital transmission depends on interactions between the parasite, the placenta, the mother, and the fetus. We review and discuss here the current knowledge about these parameters, including parasite virulence factors such as exovesicles, placental tropism, potential placental defense mechanisms, the placental transcriptome of infected women, gene polymorphism, and the maternal and fetal/neonatal immune responses, that might modulate the risk of T. cruzi congenital transmission.
Collapse
Affiliation(s)
- Ulrike Kemmerling
- Programa de Anatomía y Biología del Desarrollo, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Antonio Osuna
- Grupo de Bioquímica y Parasitología Molecular, Departamento de Parasitología, Instituto de Biotecnología, Universidad de Granada, Granada, Spain
| | - Alejandro Gabriel Schijman
- Molecular Biology of Chagas Disease Laboratory, Genetic Engineering and Molecular Biology Research Institute Dr. Héctor Torres (INGEBI-CONICET), Buenos Aires, Argentina
| | - Carine Truyens
- Laboratory of Parasitology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
49
|
Bustos PL, Milduberger N, Volta BJ, Perrone AE, Laucella SA, Bua J. Trypanosoma cruzi Infection at the Maternal-Fetal Interface: Implications of Parasite Load in the Congenital Transmission and Challenges in the Diagnosis of Infected Newborns. Front Microbiol 2019; 10:1250. [PMID: 31231337 PMCID: PMC6568191 DOI: 10.3389/fmicb.2019.01250] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 05/20/2019] [Indexed: 12/29/2022] Open
Abstract
Trypanosoma cruzi is the protozoan unicellular parasite that causes Chagas disease. It can be transmitted from infected mothers to their babies via the connatal route, thus being able to perpetuate even in the absence of Triatomine insect vectors. Chagas disease was originally endemic in Central and South America, but migration of infected women of childbearing age has spread the T. cruzi congenital infection to non-endemic areas like North America, Europe, Japan, and Australia. Currently, 7 million people are affected by this infection worldwide. This review focuses on the relevance of the T. cruzi parasite levels in different aspects of the congenital T. cruzi infection such as the mother-to-child transmission rate, the maternal and fetal immune response, and its impact on the diagnosis of infected newborns. Improvements in detection of this parasite, with tools that can be easily adapted to be used in remote rural areas, will make the early diagnosis of infected children possible, allowing a prompt trypanocidal treatment and avoiding the current loss of opportunities for the diagnosis of 100% of T. cruzi congenitally infected infants.
Collapse
Affiliation(s)
- Patricia L Bustos
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chaben" - ANLIS C. G. Malbrán, Buenos Aires, Argentina
| | - Natalia Milduberger
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chaben" - ANLIS C. G. Malbrán, Buenos Aires, Argentina.,Centro de Altos Estudios en Ciencias Humanas y de la Salud (CAECIHS), Universidad Abierta Interamericana, Buenos Aires, Argentina
| | - Bibiana J Volta
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chaben" - ANLIS C. G. Malbrán, Buenos Aires, Argentina
| | - Alina E Perrone
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chaben" - ANLIS C. G. Malbrán, Buenos Aires, Argentina
| | - Susana A Laucella
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chaben" - ANLIS C. G. Malbrán, Buenos Aires, Argentina
| | - Jacqueline Bua
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chaben" - ANLIS C. G. Malbrán, Buenos Aires, Argentina.,Centro de Altos Estudios en Ciencias Humanas y de la Salud (CAECIHS), Universidad Abierta Interamericana, Buenos Aires, Argentina
| |
Collapse
|
50
|
Espinoza B, Martínez I, Schabib-Hany M. First report of family clusters of Chagas disease seropositive blood donors in Mexico City and their epidemiological relevance. Acta Trop 2019; 193:23-30. [PMID: 30771283 DOI: 10.1016/j.actatropica.2019.02.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/06/2019] [Accepted: 02/12/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Chagas disease is an important health problem in Latin America. Relatives of T. cruzi seropositive donors could also test positive in serological assays. Therefore, the study of Chagas diseases in family clusters has become important to accurately evaluate the problem that this infectious disease represents. OBJECTIVE to investigate family cluster from blood donors, their serological, clinical and epidemiological status. METHODS 53 family clusters consisting of index case and a variable number of relatives were studied. All the participants had ELISA and Western blot assays, as well as, clinical tests including an electrocardiogram and chest x ray. RESULTS We found that 24.52% of the family clusters had at least one T. cruzi seropositive family member, in addition to the blood donor. Importantly, 20.75% of the index cases and 5.0% of the relatives presented pathological manifestations associated to Chagas disease. Several epidemiological conditions are associated to being T. cruzi seropositive. CONCLUSION blood donor's family clusters have several seropositive to T. cruzi members. Mother-child pairs were also seropositive, suggesting vertical transmition. Pathological symptom associated to Chagas Diseases were present in index cases and family member. These results highlight the importance of studying family clusters to clarify the true magnitude of Chagas disease in Mexico.
Collapse
|