1
|
Reifler K, Campbell JI, Barnett ED, Bourque DL, Hamer DH, Samra H, Camelo I. Diagnosing Chagas in Pregnancy and Childhood: What's Old and New. Clin Lab Med 2025; 45:73-86. [PMID: 39892939 DOI: 10.1016/j.cll.2024.10.004] [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] [Indexed: 02/04/2025]
Abstract
Trypanosoma cruzi, the parasite that causes Chagas disease, affects as many as 300,000 individuals in the United States. Screening individuals of childbearing age can prevent congenital transmission, and diagnosing infants and children can prevent long-term sequelae of Chagas disease. Current methods to diagnose Chagas have limations, including relying on identification of at-risk individuals and requiring at least 2 separate tests to confirm infection.
Collapse
Affiliation(s)
- Katherine Reifler
- Section of Infectious Diseases, Boston University Chobanian & Avedisian School of Medicine, Crosstown 2nd Floor, 801 Massachusetts Avenue, Boston, MA 02118, USA
| | - Jeffrey I Campbell
- Boston University Chobanian & Avedisian School of Medicine, 670 Albany Street, 6th Floor, Boston, MA 02118, USA; Section of Pediatric Infectious Diseases, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, 670 Albany Street Room 625, Boston, MA 02118, USA.
| | - Elizabeth D Barnett
- Section of Pediatric Infectious Diseases, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, 670 Albany Street Room 625, Boston, MA 02118, USA
| | - Daniel L Bourque
- Section of Infectious Diseases, Boston University Chobanian & Avedisian School of Medicine, Crosstown 2nd Floor, 801 Massachusetts Avenue, Boston, MA 02118, USA
| | - Davidson H Hamer
- Boston University School of Public Health and Chobanian & Avedisian School of Medicine, Crosstown 3rd Floor, 801 Massachusetts Avenue, Boston, MA 02118, USA
| | - Hasan Samra
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street BA 1666, Augusta, GA 30912, USA
| | - Ingrid Camelo
- Division of Pediatric Infectious Diseases, Medical College of Georgia at Augusta University, 1120 15th Street BG 2105, Augusta, GA 30912, USA
| |
Collapse
|
2
|
Cardinal MV, Enriquez GF, Gaspe MS, Fernández MDP, Capello V, Gürtler RE. Estimation of Trypanosoma cruzi infection in the main vector Triatoma infestans: accounting for imperfect detection using site-occupancy models. Parasit Vectors 2025; 18:58. [PMID: 39966871 PMCID: PMC11834302 DOI: 10.1186/s13071-025-06693-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 01/29/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Vector infection prevalence is a key component of vectorial capacity and transmission risk. Optical microscopy observation (OM) of fecal drops has been the classic method for detecting Trypanosoma cruzi infection in triatomine bugs until the advent of polymerase chain reaction (PCR)-based techniques. However, agreement among OM- and PCR-based techniques has been highly heterogeneous. METHODS We used hierarchical site-occupancy models accounting for imperfect detection to estimate method-specific detection probabilities of T. cruzi infection in field-collected Triatoma infestans and to assess whether T. cruzi infection varied with triatomine developmental stage and collection ecotope. We also performed a scoping review of the literature on comparisons between OM and PCR for T. cruzi infection diagnosis in triatomines. Triatomines were collected before vector control interventions in Pampa del Indio houses (Argentine Chaco) and examined by OM. We amplified the variable regions of the kinetoplastid minicircle genome (vkDNA-PCR) in DNA extracted from the rectal ampoules of 64 OM-positive and 65 OM-negative T. infestans. RESULTS vkDNA-PCR detected T. cruzi infection in 59 (92.2%) OM-positive bugs and in 19 (29.2%) OM-negative triatomines in blind tests. The overall prevalence of infection, as determined by a positive test result by either vkDNA-PCR or OM, was 64.3% [95% confidence interval (95% CI) 55.8-72.1%]. Detection probability of T. cruzi infection by vkDNA-PCR (92%, 95% CI 83-97%) was substantially higher than for OM (76%, 95% CI 65-84%). Infection was minimal (26.2%) in peridomestic nymphs and maximal in domestic adult triatomines (81.7%). In the literature review encompassing 26 triatomine species from 11 countries, inter-method agreement ranged from 28.6% to 100%. The lowest agreement was observed in Rhodnius sp. and Panstrongylus lutzi and the highest among Triatoma sp., with wide variability in the protocols and outcomes of molecular diagnosis in comparison with OM. CONCLUSIONS Our study provides a synthesis on the different sources (both biological and technical) of variation of the outcomes of OM- and PCR-based diagnosis of T. cruzi infection in triatomines and identifies new research needs for diagnostic improvement.
Collapse
Affiliation(s)
- Marta Victoria Cardinal
- Facultad de Ciencias Exactas y Naturales, Laboratorio de Eco-Epidemiología, Universidad de Buenos Aires, Int Güiraldes 2160, 2do piso, Ciudad Universitaria, C1428EHA, Buenos Aires, Argentina.
- CONICET-Universidad de Buenos Aires, Instituto de Ecología, Genética y Evolución de Buenos Aires (IEGEBA), Ciudad Universitaria, Buenos Aires, Argentina.
| | - Gustavo Fabián Enriquez
- Facultad de Ciencias Exactas y Naturales, Laboratorio de Eco-Epidemiología, Universidad de Buenos Aires, Int Güiraldes 2160, 2do piso, Ciudad Universitaria, C1428EHA, Buenos Aires, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Ecología, Genética y Evolución de Buenos Aires (IEGEBA), Ciudad Universitaria, Buenos Aires, Argentina
| | - María Sol Gaspe
- Facultad de Ciencias Exactas y Naturales, Laboratorio de Eco-Epidemiología, Universidad de Buenos Aires, Int Güiraldes 2160, 2do piso, Ciudad Universitaria, C1428EHA, Buenos Aires, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Ecología, Genética y Evolución de Buenos Aires (IEGEBA), Ciudad Universitaria, Buenos Aires, Argentina
| | | | - Victoria Capello
- Facultad de Ciencias Exactas y Naturales, Laboratorio de Eco-Epidemiología, Universidad de Buenos Aires, Int Güiraldes 2160, 2do piso, Ciudad Universitaria, C1428EHA, Buenos Aires, Argentina
| | - Ricardo Esteban Gürtler
- Facultad de Ciencias Exactas y Naturales, Laboratorio de Eco-Epidemiología, Universidad de Buenos Aires, Int Güiraldes 2160, 2do piso, Ciudad Universitaria, C1428EHA, Buenos Aires, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Ecología, Genética y Evolución de Buenos Aires (IEGEBA), Ciudad Universitaria, Buenos Aires, Argentina
| |
Collapse
|
3
|
Cáceres TM, Patiño LH, Ramírez JD. Understanding Host-Pathogen Interactions in Congenital Chagas Disease Through Transcriptomic Approaches. Pathogens 2025; 14:106. [PMID: 40005483 PMCID: PMC11858232 DOI: 10.3390/pathogens14020106] [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: 12/16/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
Chagas disease, caused by Trypanosoma cruzi, is a parasitic zoonosis with significant health impacts, particularly in Latin America. While traditionally associated with vector-borne transmission, increased migration has expanded its reach into urban and non-endemic regions. Congenital transmission has become a critical route of infection, involving intricate maternal-fetal immune interactions that challenge diagnosis and treatment. This review synthesizes findings from three RNA-seq studies that explore the molecular underpinnings of congenital Chagas disease, emphasizing differentially expressed genes (DEGs) implicated in host-pathogen interactions. The DAVID tool analysis highlighted the overexpression of genes associated with the innate immune response, including pro-inflammatory cytokines that drive chemotaxis and neutrophil activation. Additionally, calcium-dependent pathways critical for parasite invasion were modulated. T. cruzi exploits the maternal-fetal immune axis to establish a tolerogenic environment conducive to congenital transmission. Alterations in placental angiogenesis, cellular regeneration, and metabolic processes further demonstrate the parasite's ability to manipulate host responses for its survival and persistence. These findings underscore the complex interplay between the host and pathogen that facilitates disease progression. Future research integrating transcriptomic, proteomic, and metabolomic approaches is essential to unravel the molecular mechanisms underlying congenital Chagas disease, with a particular focus on the contributions of genetic diversity and non-coding RNAs in immune evasion and disease pathogenesis.
Collapse
Affiliation(s)
- Tatiana M. Cáceres
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá 111221, Colombia; (T.M.C.); (L.H.P.)
| | - Luz Helena Patiño
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá 111221, Colombia; (T.M.C.); (L.H.P.)
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá 111221, Colombia; (T.M.C.); (L.H.P.)
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
4
|
Campos-Ruiz M, Wang-Wang JH, Rivaya B, Rodriguez-Ponga B, López N, González V, Cardona PJ, Fernández-Rivas G. Evaluation of the Vitros® Anti-T. cruzi assay for its incorporation into the Trypanosoma cruzi infection diagnostic algorithm. Heliyon 2024; 10:e40673. [PMID: 39717580 PMCID: PMC11664266 DOI: 10.1016/j.heliyon.2024.e40673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/25/2024] Open
Abstract
Background and objective Serological screening for Chagas disease (CD) in Latin American adults living in Europe is a cost-effective strategy for transmission prevention. The World Health Organization recommends two different serological tests including native and recombinant antigens for IgG detection. In Spain, most commercialized native tests require manual processing. We aimed to evaluate the sensitivity and specificity of the automated Vitros® Anti-T. cruzi native antigen-based test. Methods A total of 556 serum samples were tested using two different tests: 1) our reference assay, a chemiluminescence immunoassay employing a recombinant multi-antigen protein (Liaison® XL Murex Chagas); 2) a chemiluminescence immunoassay with native antigen (Vitros® Anti-T. cruzi assay). Additionally, 180 samples were also processed by a manual indirect immunofluorescent assay (Chagas IFA). Sensibility, specificity and kappa index were calculated. Results Vitros® showed a kappa index of 0.94 (IC 95 %: 0.86-1.03) compared to Liaison® XL with a sensitivity of 93.6 % and specificity of 99.5 %. Compared to IFA, Vitros® showed a kappa index of 0.61 (IC 95 %: 0.47-0.76), sensitivity of 97.5 % and specificity of 70.37 %. Discrepant results were obtained mainly in treated patients. Conclusions The Vitros® Anti-T. cruzi assay showed potential for implementation as an automated serological screening test, enhancing the diagnostic process in high-throughput microbiology laboratories.
Collapse
Affiliation(s)
- Miriam Campos-Ruiz
- Microbiology Department, Clinical Laboratory North Metropolitan Area, Germans Trias I Pujol University Hospital, 08916 Badalona, Spain
- Department of Genetics and Microbiology, Autonomous University of Barcelona, 08916 Badalona, Spain
| | - Jun Hao Wang-Wang
- Microbiology Department, Clinical Laboratory North Metropolitan Area, Germans Trias I Pujol University Hospital, 08916 Badalona, Spain
- Department of Genetics and Microbiology, Autonomous University of Barcelona, 08916 Badalona, Spain
| | - Belén Rivaya
- Microbiology Department, Clinical Laboratory North Metropolitan Area, Germans Trias I Pujol University Hospital, 08916 Badalona, Spain
- Department of Genetics and Microbiology, Autonomous University of Barcelona, 08916 Badalona, Spain
| | - Beatriz Rodriguez-Ponga
- Microbiology Department, Clinical Laboratory North Metropolitan Area, Germans Trias I Pujol University Hospital, 08916 Badalona, Spain
| | - Noelia López
- Microbiology Department, Clinical Laboratory North Metropolitan Area, Germans Trias I Pujol University Hospital, 08916 Badalona, Spain
| | - Victoria González
- Microbiology Department, Clinical Laboratory North Metropolitan Area, Germans Trias I Pujol University Hospital, 08916 Badalona, Spain
- Department of Genetics and Microbiology, Autonomous University of Barcelona, 08916 Badalona, Spain
- Center for Epidemiological Studies on HIV/AIDS and STI of Catalonia (CEEISCAT), Generalitat de Catalunya, Ctra, Del Canyet, S/N, 08916 Badalona, Spain
| | - Pere-Joan Cardona
- Microbiology Department, Clinical Laboratory North Metropolitan Area, Germans Trias I Pujol University Hospital, 08916 Badalona, Spain
- Department of Genetics and Microbiology, Autonomous University of Barcelona, 08916 Badalona, Spain
| | - Gema Fernández-Rivas
- Microbiology Department, Clinical Laboratory North Metropolitan Area, Germans Trias I Pujol University Hospital, 08916 Badalona, Spain
- Department of Genetics and Microbiology, Autonomous University of Barcelona, 08916 Badalona, Spain
| |
Collapse
|
5
|
Ossowski MS, Gallardo JP, Chadi R, Hernández Y, Fernández M, Marco JD, Triana-Chavez O, Nolan MS, Pech May A, Ramsey JM, Villar JC, Agüero F, Potenza M, Gómez KA. Performance Assessment of ELISA Using the Trypanosoma cruzi-Specific Antigen Tc323 for the Diagnosis of Chronic Chagas Disease. ACS Infect Dis 2024; 10:4400-4412. [PMID: 39548973 DOI: 10.1021/acsinfecdis.4c00784] [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] [Indexed: 11/18/2024]
Abstract
In the chronic phase of Chagas disease (CCD), diagnosis relies on detecting specific IgG antibodies due to the low or absent presence of the parasiteTrypanosoma cruzi in human blood. However, the performance of current serological tests is highly variable, lacking a "gold standard" assay with 100% sensitivity and specificity, which challenges the exploration of new biomarkers. In the present study, we evaluated the diagnostic accuracy of an optimized ELISA using the predicted immunogenic domains (called TcD3 and TcD6) of Tc323, a protein highly conserved among T. cruzi strains but absent in other clinically significant parasites such as Leishmania spp. This study was conducted using plasma or serum samples from CCD individuals with different clinical manifestations and living in endemic regions in Latin America, subjects with unrelated infectious diseases, and noninfected donors. The sensitivity and specificity of recombinant TcD3 were 90.8% and 92.6%, respectively, while rTcD6 displayed values of 93.1% and 93.6% for the same parameters. Area under curve (AUC) values were 0.949 for rTcD3 and 0.954 for rTcD6. The receiver operative characteristic (ROC) curve showed a highly significant difference between CCD individuals and noninfected donors. Cross-reactivity was 10.2% for rTcD3 and 8.2% for rTcD6 in subjects infected with leishmaniasis or with toxoplasmosis. In addition, the reactivity against rTcD3 differed among some geographical areas while no significant difference was found using both domains for the detection of T. cruzi-infected individuals with or without cardiac symptoms. Our findings show that the recombinant antigens rTcD3 and rTcD6 could be used as highly potential biomarkers for the serological diagnosis of CCD.
Collapse
Affiliation(s)
- Micaela Soledad Ossowski
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI-CONICET), C1428ADN Buenos Aires, Argentina
| | - Juan Pablo Gallardo
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI-CONICET), C1428ADN Buenos Aires, Argentina
| | - Raul Chadi
- Hospital General de Agudos "Dr. Ignacio Pirovano", C1430BKC Buenos Aires, Argentina
| | - Yolanda Hernández
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chabén", C1097 Buenos Aires, Argentina
| | - Marisa Fernández
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chabén", C1097 Buenos Aires, Argentina
| | - Jorge Diego Marco
- Instituto de Investigaciones de Enfermedades Tropicales, Universidad Nacional de Salta, A4400 Orán, Argentina
- Instituto de Patología Experimental, CONICET, U9120ACD Salta, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), U9120ACD Salta, Argentina
| | - Omar Triana-Chavez
- Grupo Biología y Control de Enfermedades Infecciosas, Corporación de Patologías Tropicales, Instituto de Biología, Universidad de Antioquia, 050010 Medellin, Colombia
| | - Melissa S Nolan
- Laboratory of Vector-borne and Zoonotic Diseases, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Angelica Pech May
- Instituto Nacional de Medicina Tropical (INMET-ANLIS), Ministerio de Salud de la Nación, Puerto Iguazu, C1073ABA Misiones, Argentina
| | - Janine M Ramsey
- Centro Regional de Investigación en Salud Pública, Instituto Nacional de Salud Pública, Tapachula 62100, México
| | - Juan C Villar
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga, Santander 680003, Colombia
- Fundación Cardioinfantil - Instituto de Cardiología, Bogota 14780110, Colombia
| | - Fernán Agüero
- Instituto de Investigaciones Biotecnológicas (IIB) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martin, B1650HMQ Buenos Aires, Argentina
- Escuela de Bio y Nanotecnología (EByN), Universidad Nacional de San Martín, San Martin, A4419XAP Buenos Aires, Argentina
| | - Mariana Potenza
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI-CONICET), C1428ADN Buenos Aires, Argentina
| | - Karina Andrea Gómez
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI-CONICET), C1428ADN Buenos Aires, Argentina
| |
Collapse
|
6
|
Tabares-Medina J, García-Blandón K, García-Montoya GM, Soto-Calderón ID. Redefining infections with trypanosomatids in Neotropical primates: Case study of the white-footed tamarin ( Oedipomidas leucopus). Int J Parasitol Parasites Wildl 2024; 25:101021. [PMID: 39640376 PMCID: PMC11617692 DOI: 10.1016/j.ijppaw.2024.101021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/12/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024]
Abstract
Trypanosomes are blood parasites capable of infecting nearly any vertebrate. Many Neotropical primates frequently host trypanosomes and are considered potential reservoirs for Trypanosoma cruzi and other human-pathogenic trypanosomatids. However, diagnostic methods originally developed for detecting these trypanosomatids in humans and domestic species must be validated to reliably diagnose infections in non-human primates. Without such validation, taxonomic biases and incorrect assignments of wildlife reservoirs can occur. The white-footed tamarin (Oedipomidas leucopus), a primate endemic to northwestern Colombia, is classified by the World Health Organization as a reservoir of T. cruzi. However, this classification is based on studies with small sample sizes, ambiguous diagnostic methods, and questionable geographic records. In this study, the 18S ribosomal RNA gene was amplified via PCR and sequenced to estimate trypanosome infection rates and identify species in natural populations of O. leucopus across a wide geographic range, as well as in (ex situ) specimens. This molecular approach was also compared with traditional microscopy diagnosis using blood smears. The molecular diagnosis revealed that over 60% of the tested specimens were infected, whereas traditional microscopy resulted in 58% false negatives compared to the molecular method. A Bayesian phylogeny of the 18S gene identified T. minasense as the sole trypanosomatid species present in O. leucopus, with no detections of T. cruzi or other trypanosomatids of concern to human or domestic animal health. This study highlights the risk of overestimating the presence of human-infecting trypanosomes, such as T. cruzi, in tamarins and other vertebrates, and underscores the importance of validating diagnostic methods to accurately assess the zoonotic potential of wild species. Accurate identification of wildlife reservoirs is essential for understanding parasite life cycles and implementing effective management and conservation strategies for primates and other potential reservoirs.
Collapse
Affiliation(s)
- Juliana Tabares-Medina
- Laboratorio de Genética Animal. Grupo Agrociencias, Biodiversidad y Territorio, Instituto de Biología, Facultad de Ciencias Exactas y Naturales, Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Katherinne García-Blandón
- Laboratorio de Genética Animal. Grupo Agrociencias, Biodiversidad y Territorio, Instituto de Biología, Facultad de Ciencias Exactas y Naturales, Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Gisela M. García-Montoya
- Centro Nacional de Secuenciación Genómica-CNSG, Sede de Investigación Universitaria-SIU, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia, Medellín, Antioquia, Colombia
- Grupo de Microbióloga ambiental. Escuela de Microbiología, Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Iván Darío Soto-Calderón
- Laboratorio de Genética Animal. Grupo Agrociencias, Biodiversidad y Territorio, Instituto de Biología, Facultad de Ciencias Exactas y Naturales, Universidad de Antioquia, Medellín, Antioquia, Colombia
| |
Collapse
|
7
|
Reis-Cunha JL, Jeffares DC. Detecting complex infections in trypanosomatids using whole genome sequencing. BMC Genomics 2024; 25:1011. [PMID: 39472783 PMCID: PMC11520695 DOI: 10.1186/s12864-024-10862-6] [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: 06/27/2024] [Accepted: 10/03/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Trypanosomatid parasites are a group of protozoans that cause devastating diseases that disproportionately affect developing countries. These protozoans have developed several mechanisms for adaptation to survive in the mammalian host, such as extensive expansion of multigene families enrolled in host-parasite interaction, adaptation to invade and modulate host cells, and the presence of aneuploidy and polyploidy. Two mechanisms might result in "complex" isolates, with more than two haplotypes being present in a single sample: multiplicity of infections (MOI) and polyploidy. We have developed and validated a methodology to identify multiclonal infections and polyploidy using whole genome sequencing reads, based on fluctuations in allelic read depth in heterozygous positions, which can be easily implemented in experiments sequencing genomes from one sample to larger population surveys. RESULTS The methodology estimates the complexity index (CI) of an isolate, and compares real samples with simulated clonal infections at individual and populational level, excluding regions with somy and gene copy number variation. It was primarily validated with simulated MOI and known polyploid isolates respectively from Leishmania and Trypanosoma cruzi. Then, the approach was used to assess the complexity of infection using genome wide SNP data from 497 trypanosomatid samples from four clades, L. donovani/L. infantum, L. braziliensis, T. cruzi and T. brucei providing an overview of multiclonal infection and polyploidy in these cultured parasites. We show that our method robustly detects complex infections in samples with at least 25x coverage, 100 heterozygous SNPs and where 5-10% of the reads correspond to the secondary clone. We find that relatively small proportions (≤ 7%) of cultured trypanosomatid isolates are complex. CONCLUSIONS The method can accurately identify polyploid isolates, and can identify multiclonal infections in scenarios with sufficient genome read coverage. We pack our method in a single R script that requires only a standard variant call format (VCF) file to run ( https://github.com/jaumlrc/Complex-Infections ). Our analyses indicate that multiclonality and polyploidy do occur in all clades, but not very frequently in cultured trypanosomatids. We caution that our estimates are lower bounds due to the limitations of current laboratory and bioinformatic methods.
Collapse
Affiliation(s)
- João Luís Reis-Cunha
- York Biomedical Research Institute, Department of Biology and York Biomedical Research Institute, University of York, York, YO10 5DD, UK.
| | - Daniel Charlton Jeffares
- York Biomedical Research Institute, Department of Biology and York Biomedical Research Institute, University of York, York, YO10 5DD, UK.
| |
Collapse
|
8
|
Swett MC, Rayes DL, Campos SV, Kumar RN. Chagas Disease: Epidemiology, Diagnosis, and Treatment. Curr Cardiol Rep 2024; 26:1105-1112. [PMID: 39115799 DOI: 10.1007/s11886-024-02113-7] [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] [Accepted: 08/01/2024] [Indexed: 10/09/2024]
Abstract
PURPOSE OF REVIEW This review seeks to describe the updates in the literature - particularly with regards to the epidemiology and diagnosis of Chagas disease. Additionally, this paper describes updates to the antiparasitic treatment for Chagas disease. RECENT FINDINGS With regards to changing epidemiology, autochthonous cases are being found within the USA in addition to Latin America. Additionally, there appears to be more intermixing of discrete typing units-meaning, they are not confined to specific geographic regions. Screening for Chagas disease is recommended in persons who lived in areas with endemic Chagas, persons wtih family member diagnosed with Chagas Disease, persons who have lived in homes of natural material in Latin America, and persons with history of kissing bug bites. Treatment for the parasitic infection remains limited to benznidazole and nifurtimox, and the role of these treatments in Chagas cardiomyopathy has not yet been definitively defined. Finally, indications for and management of heart transplant in the setting of Chagas disease are discussed. FUTURE RESEARCH Use of antiparasitics during chronic chagas disease should be further explored. Additionally, future research identifying other markers of infection would be valuable to defining cure from infection.
Collapse
Affiliation(s)
- Michael C Swett
- Department of Internal Medicine, MedStar Georgetown University Hospital, Washington, DC, USA
| | - Danny L Rayes
- Department of Internal Medicine, MedStar Georgetown University Hospital, Washington, DC, USA
| | - Silvia Vidal Campos
- Pulmonary Division, Heart Institute (InCor), University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Rebecca N Kumar
- Division of Infectious Disease and Tropical Medicine, MedStar Georgetown University Hospital, Washington, DC, USA.
| |
Collapse
|
9
|
Xie Y, Wang J, Wang Y, Wen Y, Pu Y, Wang B. Parasite-enhanced immunotherapy: transforming the "cold" tumors to "hot" battlefields. Cell Commun Signal 2024; 22:448. [PMID: 39327550 PMCID: PMC11426008 DOI: 10.1186/s12964-024-01822-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/08/2024] [Indexed: 09/28/2024] Open
Abstract
Immunotherapy has emerged as a highly effective treatment for various tumors. However, the variable response rates associated with current immunotherapies often restrict their beneficial impact on a subset of patients. Therefore, more effective treatment approaches that can broaden the scope of therapeutic benefits to a larger patient population are urgently needed. Studies have shown that some parasites and their products, for example, Plasmodium, Toxoplasma, Trypanosoma, and Echinococcus, can effectively transform "cold" tumors into "hot" battlefields and reshape the tumor microenvironment, thereby stimulating innate and adaptive antitumor immune responses. These parasitic infections not only achieve the functional reversal of innate immune cells, such as neutrophils, macrophages, myeloid-derived suppressor cells, regulatory T cells, and dendritic cells, in tumors but also successfully activate CD4+/CD8+ T cells and even B cells to produce antibodies, ultimately resulting in an antitumor-specific immune response and antibody-dependent cellular cytotoxicity. Animal studies have confirmed these findings. This review discusses the abovementioned content and the challenges faced in the future clinical application of antitumor treatment strategies based on parasitic infections. With the potential of these parasites and their byproducts to function as anticancer agents, we anticipate that further investigations in this field could yield significant advancements in cancer treatment.
Collapse
Affiliation(s)
- Yujun Xie
- Laboratory of Tumor Immunobiology, Department of Public Health and Pathogen Biology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Graduate School, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Jinyan Wang
- College of Basic Medical Science, China Medical University, Shenyang, Liaoning, 110122, China
| | - Yafei Wang
- Faculty of Arts and Science, University of Toronto, Toronto, ON, M5S 3G3, Canada
| | - Yalin Wen
- Laboratory of Tumor Immunobiology, Department of Public Health and Pathogen Biology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Graduate School, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Yanping Pu
- Graduate School, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Benfan Wang
- Laboratory of Tumor Immunobiology, Department of Public Health and Pathogen Biology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China.
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China.
- Institute of Surgery, The First Affiliated Hospital, Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China.
| |
Collapse
|
10
|
Simões-Neto EA, Santos DWDCL, Bomfim MRQ, Costa JML, Simões AF, Vasconcelos LD, Sodré DC, Costa ACM, Dumont SVR, de Oliveira de Melo B, de Azevedo CDMPES. Oral Chagas disease outbreak by bacaba juice ingestion: A century after Carlos Chagas' discovery, the disease is still hard to manage. PLoS Negl Trop Dis 2024; 18:e0012225. [PMID: 39292716 PMCID: PMC11441692 DOI: 10.1371/journal.pntd.0012225] [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: 05/19/2024] [Revised: 09/30/2024] [Accepted: 07/24/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND Orally transmitted acute Chagas disease (ACD) primarily affects low-visibility and low-income individuals in tropical and subtropical zones. Managing ACD remains challenging even after more than 100 years of its discovery. Its spread to non-endemic areas has made it a global health issue. The aim of this work is to demonstrate the difficulties encountered in handling a real-life situation. METHODOLOGY AND FINDINGS This report examines an outbreak of 39 cases of ACD due to oral transmission by bacaba juice ingestion that occurred in Pedro do Rosário, Maranhão, Brazil. A clinical and epidemiological investigation, including an entomological search, was conducted. Diagnosis criteria included positive peripheral blood smear (PBS), seroconversion of IgG, and a two-fold increase in IgG titer (laboratory criteria); and clinical findings, epidemiological exposure, and at least one positive IgG test (clinical-epidemiological criteria). In-house conventional polymerase chain reaction (PCR) was performed on 33 samples. All patients were treated with benznidazole. After 4.5 years, IgG levels were reassessed in 26 individuals. The mean age was 33.6 years, with no gender difference. The mean incubation period was 13.8 days, and the mean between symptom onset and treatment was 16.6 days. The most common symptoms were fever and lymphadenopathy (90%). Diagnostic success rates were 66.6% (laboratory criteria), 23% (clinical-epidemiological criteria), and 10.2% (high clinical suspicion despite negative tests). Test positivity rates were 69.7% (PBS), 91.4% (serology), and 100% (PCR). There were no deaths. Serological cure was achieved in 34.6% of cases, and IgG titers decreased in 15.3%. CONCLUSIONS AND SIGNIFICANCE We encountered several barriers in managing ACD, including population vulnerability, reliance on outdated diagnostic techniques, lack of standardized molecular biology methods, and limited therapeutic options. This report underscores the importance of rapid surveillance and early treatment to prevent fatalities. We recommend the standardization of conventional PCR in diagnostic routines.
Collapse
Affiliation(s)
| | - Daniel Wagner de Castro Lima Santos
- Presidente Dutra University Hospital (HU-UPD), Brazilian Company of Hospital Services (EBSERH), Federal University of Maranhão (UFMA), São Luís, Brazil
- IDOR, Instituto D'Or de Pesquisa e Ensino
| | | | | | - Amanda Ferreira Simões
- Brasília University Hospital (HUB), Brazilian Company of Hospital Services (EBSERH), Brasília University (UnB), Brasília, Brazil
| | | | | | | | | | | | | |
Collapse
|
11
|
Ricardo-Caldera D, Espitia-Pérez L, Avilés-Vergara PA, Benítez ÁJ, Chacón-Pacheco J, Ballesteros-Correa J, Negrette-Oquendo A, Soto-De León S, Tovar-Acero C. Trypanosoma cruzi in domestic and wild mammals in the northeast region of Colombia. Int J Parasitol Parasites Wildl 2024; 24:100940. [PMID: 38708188 PMCID: PMC11067360 DOI: 10.1016/j.ijppaw.2024.100940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/07/2024]
Abstract
Trypanosoma cruzi hosts can serve as a source of infection for animals, vectors, and humans, contributing to the establishment of Chagas disease (CD) in a given area. Traditionally, the Department of Córdoba has not been considered a transmission area for CD; however, the report of several acute cases of Chagas disease highlights the importance of studying the dynamics of disease transmission in this region. This study aimed to detect T. cruzi in domestic and wild mammals in the department of Córdoba. In 2017, a cross-sectional descriptive study was conducted in six villages in two municipalities in the department of Córdoba. Blood samples from dogs living in the zones were collected in EDTA vacutainer tubes for domestic mammals. Wild mammals were collected using Sherman and Tomahawk traps and mist nets in crops and peridomiciles. T. cruzi DNA was detected using the kinetoplast DNA (kDNA) variable region and the tandem repeat satellite region of T. cruzi as molecular targets. We sampled 168 dogs and 146 wild mammals. The detected prevalence of T. cruzi was 6.37%; the TcI lineage was found in D. marsupialis, H. anomalus, and one canine. A specimen of D. marsupialis with TcI and TcII lineages was also identified. T. cruzi DNA was detected in domestic and wild animals in the study area, indicating the circulation of the parasite in peridomestic environments. D. marsupialis may represent an important host in maintaining this region's wild and domestic cycle.
Collapse
Affiliation(s)
- Dina Ricardo-Caldera
- Grupo de investigación en Enfermedades Tropicales y Resistencia Bacteriana, Facultad de Ciencias de la Salud, Universidad del Sinú, Montería, Colombia
| | - Lyda Espitia-Pérez
- Grupo de Investigación Biomédica y Biología Molecular, Facultad de Ciencias de la Salud, Universidad del Sinú, Montería, Colombia
| | - Paula A. Avilés-Vergara
- Grupo de investigación en Enfermedades Tropicales y Resistencia Bacteriana, Facultad de Ciencias de la Salud, Universidad del Sinú, Montería, Colombia
| | - Álvaro J. Benítez
- Grupo de investigación en Enfermedades Tropicales y Resistencia Bacteriana, Facultad de Ciencias de la Salud, Universidad del Sinú, Montería, Colombia
- Núcleo de Pesquisa em Ciências Biológicas & Departamento de Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| | - Julio Chacón-Pacheco
- Grupo de Investigación Biodiversidad Unicórdoba, Universidad de Córdoba, Montería, Colombia
- Laboratorio de Ecología Funcional Unidad de Ecología y Sistemática (UNESIS), Laboratorio de Ecología Funcional, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | - Ana Negrette-Oquendo
- Grupo de investigación en Enfermedades Tropicales y Resistencia Bacteriana, Facultad de Ciencias de la Salud, Universidad del Sinú, Montería, Colombia
| | - Sara Soto-De León
- Grupo de investigación en Enfermedades Tropicales y Resistencia Bacteriana, Facultad de Ciencias de la Salud, Universidad del Sinú, Montería, Colombia
| | - Catalina Tovar-Acero
- Grupo de investigación en Enfermedades Tropicales y Resistencia Bacteriana, Facultad de Ciencias de la Salud, Universidad del Sinú, Montería, Colombia
| |
Collapse
|
12
|
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
|
13
|
Davila E, Fernandez-Santos NA, Estrada-Franco JG, Wei L, Velázquez-Ramírez DD, García-Miranda R, Irecta Nájera C, Cruz-Cadena R, Guichard-Romero C, Rodriguez C, Tarleton R, Rodríguez-Pérez MA, Ochoa-Díaz-López H, Hamer GL, Hamer SA. Domestic Dog Infection with Trypanosoma cruzi from Northern and Southern Regions of Mexico. Vector Borne Zoonotic Dis 2024; 24:510-519. [PMID: 38949980 DOI: 10.1089/vbz.2023.0110] [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] [Indexed: 07/03/2024] Open
Abstract
Background: Chagas disease or American trypanosomiasis, caused by Trypanosoma cruzi and vectored by triatomines, affects millions of people worldwide. In endemic countries including Mexico, infections in domestic animals, such as dogs, may affect the risk of human disease when they serve as a source of infection to vectors that subsequently infect humans. Materials and Methods: We conducted a cross-sectional study of 296 dogs from two cities near the northern and southern borders of Mexico: Reynosa, Tamaulipas, and Tuxtla Gutierrez, Chiapas. Infection was measured based on testing of blood using T. cruzi quantitative PCR (qPCR) and up to three antibody detection assays. The StatPak immunochromatographic assay was used to screen samples and the indirect fluorescent antibody (IFA) and multiplex microsphere immunoassay (MIA) tests were used as secondary tests on all samples that screened positive and a subset of negatives. Serologic positivity was defined based on reactivity on at least two independent tests. Results: Of the 280 samples tested for parasite DNA, two (0.7%) were positive, one of which (0.4%) was confirmed as T. cruzi discrete typing unit TcIV. Overall, 72 (24.3%) samples were reactive for T. cruzi antibodies via StatPak of which 8 were also positive using MIA and 2 were also positive using IFA (including one of the PCR-positive dogs). Overall, nine dogs (3.4%) met study criteria of positivity based on either/both serology or PCR tests. Positive dogs were found in both regions of Mexico; five (2.7%) from Reynosa and four (3.6%) from Tuxtla Gutierrez. We found no association between infection status and state of origin, sex, age group, breed group, neighborhood, and whether other pets lived in the home. Conclusion: Our results re-emphasize dogs' utility as sentinels for T. cruzi in Mexico and underscore the need for improved veterinary diagnostic tests and parasite surveillance at the household level in endemic countries.
Collapse
Affiliation(s)
- Edward Davila
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, USA
| | - Nadia A Fernandez-Santos
- Department of Entomology, Texas A&M University, College Station, Texas, USA
- Instituto Politécnico Nacional, Centro de Biotecnología Genómica, Laboratorio de Biomedicina Molecular, Reynosa, Tamaulipas, Mexico
| | - José Guillermo Estrada-Franco
- Instituto Politécnico Nacional, Centro de Biotecnología Genómica, Laboratorio de Biomedicina Molecular, Reynosa, Tamaulipas, Mexico
| | - Lihua Wei
- Instituto Politécnico Nacional, Centro de Biotecnología Genómica, Laboratorio de Biomedicina Molecular, Reynosa, Tamaulipas, Mexico
| | | | - Rosario García-Miranda
- Departamento de Salud, El Colegio de la Frontera Sur (ECOSUR), San Cristóbal de Las Casas, México
- Escuela de Lenguas, Universidad Autónoma de Chiapas (UNACH), San Cristóbal de Las Casas, México
| | - Cesar Irecta Nájera
- Departamento de Salud, El Colegio de la Frontera Sur (ECOSUR), Villahermosa, México
| | - Raúl Cruz-Cadena
- Escuela de Ciencias Químicas, Universidad Autónoma de Chiapas (UNACH), Ocozocoautla de Espinosa, México
| | | | - Carlos Rodriguez
- Texas A&M Veterinary Medical Diagnostic Laboratory, College Station, Texas, USA
| | - Rick Tarleton
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| | - Mario A Rodríguez-Pérez
- Instituto Politécnico Nacional, Centro de Biotecnología Genómica, Laboratorio de Biomedicina Molecular, Reynosa, Tamaulipas, Mexico
| | - Héctor Ochoa-Díaz-López
- Departamento de Salud, El Colegio de la Frontera Sur (ECOSUR), San Cristóbal de Las Casas, México
| | - Gabriel L Hamer
- Department of Entomology, Texas A&M University, College Station, Texas, USA
| | - Sarah A Hamer
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
14
|
Cantillo-Barraza O, Jaimes-Dueñez J, Marcet PL, Triana-Chavez O, Gómez-Palacio A. Multilocus genetic analysis of Trypanosoma cruzi supports non-domestic intrusion into domestic transmission in an endemic region of Colombia. Parasite Epidemiol Control 2024; 26:e00364. [PMID: 39021811 PMCID: PMC11253143 DOI: 10.1016/j.parepi.2024.e00364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 05/27/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Trypanosoma cruzi, the causative agent of Chagas disease, is primarily transmitted to humans by hematophagous bugs of the Triatominae subfamily. In the Colombian Caribbean region, particularly on Margarita Island, T. cruzi transmission is highly endemic and associated with vectors such as Triatoma maculata and Rhodnius pallescens. Additionally, T. cruzi-infected Didelphis marsupialis are commonly found in close proximity to human dwellings. Given the complex transmission dynamics involving various domestic and non-domestic hosts, this study aimed to analyze 145 T. cruzi clones from twelve strains isolated from T. maculata, R. pallescens, and D. marsupialis using spliced leader intergenic region (SL-IR) sequences and nine polymorphic microsatellite loci. The results indicate the presence of a single polymorphic T. cruzi population, suggesting sustained local transmission dynamics between triatomines adapted to A. butyracea forests and peridomestic areas inhabited by synanthropic mammal reservoir such as D. marsupialis. Notably, this population appears to lack substructure, highlighting the importance of adopting an alternative eco-health approach to complement traditional chemical vector control methods for more effective and sustainable interruption of transmission.
Collapse
Affiliation(s)
- Omar Cantillo-Barraza
- Grupo Biología y Control de Enfermedades Infecciosas (BCEI), Universidad de Antioquia, Medellín, Colombia
| | - Jeiczon Jaimes-Dueñez
- Grupo de Investigación en Ciencias Animales (GRICA), Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia (UCC), Bucaramanga, Colombia
| | - Paula L. Marcet
- Centers for Disease Control and Prevention (CDC), Division of Parasitic Diseases and Malaria, Entomology Branch, Atlanta, GA, USA
| | - Omar Triana-Chavez
- Grupo Biología y Control de Enfermedades Infecciosas (BCEI), Universidad de Antioquia, Medellín, Colombia
| | - Andrés Gómez-Palacio
- Laboratorio de Investigación en Genética Evolutiva (LIGE), Universidad Pedagógica y Tecnológica de Colombia, Tunja, Boyacá, Colombia
| |
Collapse
|
15
|
Llovera A, Abras A, Fernández-Arévalo A, Ballart C, Heras S, Muñoz C, Gállego M. Genetic Diversity of Trypanosoma cruzi in the United States of America: The Least Endemic Country for Chagas Disease. Life (Basel) 2024; 14:901. [PMID: 39063654 PMCID: PMC11278504 DOI: 10.3390/life14070901] [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: 06/21/2024] [Revised: 07/05/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Chagas disease (CD), caused by Trypanosoma cruzi and endemic in Latin America, has become an emergent health problem in non-endemic countries due to human migration. The United States (US) is the non-Latin American country with the highest CD burden and cannot be considered as non-endemic, since triatomine vectors and reservoir animals have been found. Populations of T. cruzi are divided into genetic subdivisions, which are known as discrete typing units (DTUs): TcI to TcVI and TcBat. Autochthonous human T. cruzi infection in the US is sporadic, but it may change due to environmental factors affecting the geographic distribution of triatomines. We aimed to perform a literature review of the genetic diversity of T. cruzi in triatomine vectors and mammalian hosts, including human cases, in the US. The 34 analyzed studies revealed the presence of T. cruzi in 18 states, which was mainly concentrated in Texas, Louisiana and New Mexico. TcI and TcIV were the principal DTUs identified, being TcI the most genotyped (42.4%; 917/2164). This study represents a first attempt to compile the molecular epidemiology of T. cruzi in the US, which is fundamental for predicting the progression of the infection in the country and could be of great help in its future management.
Collapse
Affiliation(s)
| | - Alba Abras
- Laboratori d’Ictiologia Genètica, Departament de Biologia, Universitat de Girona, 17003 Girona, Spain;
| | - Anna Fernández-Arévalo
- Secció de Parasitologia, Departament de Biologia, Sanitat i Medi Ambient, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain; (A.F.-A.); (C.B.); (M.G.)
| | - Cristina Ballart
- Secció de Parasitologia, Departament de Biologia, Sanitat i Medi Ambient, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain; (A.F.-A.); (C.B.); (M.G.)
- Institut de Salut Global de Barcelona (ISGlobal), Hospital Clínic, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Sandra Heras
- Laboratori d’Ictiologia Genètica, Departament de Biologia, Universitat de Girona, 17003 Girona, Spain;
| | - Carmen Muñoz
- Servei de Microbiologia, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain;
- Institut de Recerca Biomèdica Sant Pau, 08041 Barcelona, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Montserrat Gállego
- Secció de Parasitologia, Departament de Biologia, Sanitat i Medi Ambient, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain; (A.F.-A.); (C.B.); (M.G.)
- Institut de Salut Global de Barcelona (ISGlobal), Hospital Clínic, Universitat de Barcelona, 08036 Barcelona, Spain
- CIBERINFEC (Centro de Investigación Biomédica en Red de Enfermedades Infecciosas), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
16
|
Rossi F, Santonicola S, Amadoro C, Marino L, Colavita G. Food and Drinking Water as Sources of Pathogenic Protozoans: An Update. APPLIED SCIENCES 2024; 14:5339. [DOI: 10.3390/app14125339] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
This narrative review was aimed at collecting updated knowledge on the risk factors, illnesses caused, and measures for the prevention of protozoan infections transmitted by food and drinking water. Reports screened dated from 2019 to the present and regarded global prevalence in food handlers, occurrence in food and drinking water, impact on human health, and recently reported outbreaks and cases of severe infections attributable to the dietary route. Cryptosporidium spp., Cyclospora cayetanensis, Entamoeba histolytica, and Cystoisospora belli were the protozoans most frequently involved in recently reported waterborne and foodborne outbreaks and cases. Blastocystis hominis was reported to be the most widespread intestinal protozoan in humans, and two case reports indicated its pathogenic potential. Dientamoeba fragilis, Endolimax nana, and Pentatrichomonas hominis are also frequent but still require further investigation on their ability to cause illness. A progressive improvement in surveillance of protozoan infections and infection sources took place in developed countries where the implementation of reporting systems and the application of molecular diagnostic methods led to an enhanced capacity to identify epidemiological links and improve the prevention of foodborne and waterborne protozoan infections.
Collapse
Affiliation(s)
- Franca Rossi
- Istituto Zooprofilattico Sperimentale dell’Abruzzo e Molise (IZSAM), 86100 Campobasso, Italy
| | - Serena Santonicola
- Dipartimento di Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy
| | - Carmela Amadoro
- Dipartimento di Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy
| | - Lucio Marino
- Istituto Zooprofilattico Sperimentale dell’Abruzzo e Molise (IZSAM), 86100 Campobasso, Italy
| | - Giampaolo Colavita
- Dipartimento di Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy
| |
Collapse
|
17
|
Maldonado E, Canobra P, Oyarce M, Urbina F, Miralles VJ, Tapia JC, Castillo C, Solari A. In Vitro Identification of Phosphorylation Sites on TcPolβ by Protein Kinases TcCK1, TcCK2, TcAUK1, and TcPKC1 and Effect of Phorbol Ester on Activation by TcPKC of TcPolβ in Trypanosoma cruzi Epimastigotes. Microorganisms 2024; 12:907. [PMID: 38792752 PMCID: PMC11124317 DOI: 10.3390/microorganisms12050907] [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: 03/01/2024] [Revised: 04/06/2024] [Accepted: 04/17/2024] [Indexed: 05/26/2024] Open
Abstract
Chagas disease is caused by the single-flagellated protozoan Trypanosoma cruzi, which affects several million people worldwide. Understanding the signal transduction pathways involved in this parasite's growth, adaptation, and differentiation is crucial. Understanding the basic mechanisms of signal transduction in T. cruzi could help to develop new drugs to treat the disease caused by these protozoa. In the present work, we have demonstrated that Fetal Calf Serum (FCS) can quickly increase the levels of both phosphorylated and unphosphorylated forms of T. cruzi DNA polymerase beta (TcPolβ) in tissue-cultured trypomastigotes. The in vitro phosphorylation sites on TcPolβ by protein kinases TcCK1, TcCK2, TcAUK1, and TcPKC1 have been identified by Mass Spectrometry (MS) analysis and with antibodies against phosphor Ser-Thr-Tyr. MS analysis indicated that these protein kinases can phosphorylate Ser and Thr residues on several sites on TcPolβ. Unexpectedly, it was found that TcCK1 and TcPKC1 can phosphorylate a different Tyr residue on TcPolβ. By using a specific anti-phosphor Tyr monoclonal antibody, it was determined that TcCK1 can be in vitro autophosphorylated on Tyr residues. In vitro and in vivo studies showed that phorbol 12-myristate 13-acetate (PMA) can activate the PKC to stimulate the TcPolβ phosphorylation and enzymatic activity in T. cruzi epimastigotes.
Collapse
Affiliation(s)
- Edio Maldonado
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (P.C.); (M.O.); (F.U.); (J.C.T.)
| | - Paz Canobra
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (P.C.); (M.O.); (F.U.); (J.C.T.)
| | - Matías Oyarce
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (P.C.); (M.O.); (F.U.); (J.C.T.)
| | - Fabiola Urbina
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (P.C.); (M.O.); (F.U.); (J.C.T.)
| | - Vicente J. Miralles
- Departamento de Bioquímica y Biología Molecular, Universidad de Valencia, 46110 Valencia, Spain;
| | - Julio C. Tapia
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (P.C.); (M.O.); (F.U.); (J.C.T.)
| | - Christian Castillo
- Programa de Anatomía y Biología del Desarrollo, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile;
| | - Aldo Solari
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (P.C.); (M.O.); (F.U.); (J.C.T.)
| |
Collapse
|
18
|
Ascanio LC, Carroll S, Paniz-Mondolfi A, Ramírez JD. In vitro diagnostic methods of Chagas disease in the clinical laboratory: a scoping review. Front Microbiol 2024; 15:1393992. [PMID: 38746745 PMCID: PMC11091413 DOI: 10.3389/fmicb.2024.1393992] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/02/2024] [Indexed: 01/31/2025] Open
Abstract
Background Chagas disease (CD), caused by Trypanosoma cruzi, is a global health concern with expanding geographical reach. Despite improved and accessible test methods, diagnosing CD in its various phases remains complex. The existence of clinical scenarios, including immunosuppressed patients, transplant-related CD reactivation, transfusion-associated cases, and orally transmitted acute infections, adds to the diagnostic challenge. No singular gold standard test exists for all phases, and recommendations from PAHO and the CDC advocate for the use of two serological methods for chronic CD diagnosis, while molecular methods or direct parasite detection are suggested for the acute phase. Given the complexity in the diagnostic landscape of CD, the goal of this scoping review is to characterize available diagnostic tests for CD in the clinical laboratory. Methods A literature search in PubMed was conducted on studies related to In vitro diagnosis (IVD) in humans published in English, Spanish, or Portuguese language as of 28 August 2023, and extended backward with no predefined time frame. Studies underwent title and abstract screening, followed by full-text review. Studies included were classified based on the diagnostic method used. Test methods were grouped as serological, molecular, and other methods. Performance, availability, and regulatory status were also characterized. Results Out of 85 studies included in the final review, 115 different tests were identified. These tests comprised 89 serological test types, 21 molecular test types, and 5 other test methods. Predominant serological tests included ELISA (38 studies, 44.70%), Rapid tests (19 studies, 22.35%), and chemiluminescence (10 studies, 11.76%). Among molecular tests, Polymerase Chain Reaction (PCR) assays were notable. Twenty-eight tests were approved globally for IVD or donor testing, all being serological methods. Molecular assays lacked approval for IVD in the United States, with only European and Colombian regulatory acceptance. Discussion and conclusion Serological tests, specifically ELISAs, remain the most used and commercially available diagnostic methods. This makes sense considering that most Chagas disease diagnoses occur in the chronic phase and that the WHO gold standard relies on 2 serological tests to establish the diagnosis of chronic Chagas. ELISAs are feasible and relatively low-cost, with good performance with sensitivities ranging between 77.4% and 100%, and with specificities ranging between 84.2% and 100%. Molecular methods allow the detection of specific variants but rely on the parasite's presence, which limits their utility to parasitemia levels. Depending on the PCR method and the phase of the disease, the sensitivity ranged from 58.88 to 100% while the mean specificity ranged from 68.8% to 100%. Despite their performance, molecular testing remains mostly unavailable for IVD use. Only 3 molecular tests are approved for IVD, which are available only in Europe. Six commercial serological assays approved by the FDA are available for blood and organ donor screening. Currently, there are no guidelines for testing CD oral outbreaks. Although more evidence is needed on how testing methods should be used in special clinical scenarios, a comprehensive approach of clinical assessment and diagnostics tests, including not IVD methods, is required for an accurate CD diagnosis.
Collapse
Affiliation(s)
- Luis C. Ascanio
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Savannah Carroll
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Alberto Paniz-Mondolfi
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Juan David Ramírez
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
19
|
Schaub GA. Interaction of Trypanosoma cruzi, Triatomines and the Microbiota of the Vectors-A Review. Microorganisms 2024; 12:855. [PMID: 38792688 PMCID: PMC11123833 DOI: 10.3390/microorganisms12050855] [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: 03/03/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 05/26/2024] Open
Abstract
This review summarizes the interactions between Trypanosoma cruzi, the etiologic agent of Chagas disease, its vectors, triatomines, and the diverse intestinal microbiota of triatomines, which includes mutualistic symbionts, and highlights open questions. T. cruzi strains show great biological heterogeneity in their development and their interactions. Triatomines differ from other important vectors of diseases in their ontogeny and the enzymes used to digest blood. Many different bacteria colonize the intestinal tract of triatomines, but only Actinomycetales have been identified as mutualistic symbionts. Effects of the vector on T. cruzi are indicated by differences in the ability of T. cruzi to establish in the triatomines and in colonization peculiarities, i.e., proliferation mainly in the posterior midgut and rectum and preferential transformation into infectious metacyclic trypomastigotes in the rectum. In addition, certain forms of T. cruzi develop after feeding and during starvation of triatomines. Negative effects of T. cruzi on the triatomine vectors appear to be particularly evident when the triatomines are stressed and depend on the T. cruzi strain. Effects on the intestinal immunity of the triatomines are induced by ingested blood-stage trypomastigotes of T. cruzi and affect the populations of many non-symbiotic intestinal bacteria, but not all and not the mutualistic symbionts. After the knockdown of antimicrobial peptides, the number of non-symbiotic bacteria increases and the number of T. cruzi decreases. Presumably, in long-term infections, intestinal immunity is suppressed, which supports the growth of specific bacteria, depending on the strain of T. cruzi. These interactions may provide an approach to disrupt T. cruzi transmission.
Collapse
Affiliation(s)
- Günter A Schaub
- Zoology/Parasitology, Ruhr-University Bochum, Universitätsstr. 150, 44780 Bochum, Germany
| |
Collapse
|
20
|
Cáceres TM, Cruz-Saavedra L, Patiño LH, Ramírez JD. Comparative analysis of metacyclogenesis and infection curves in different discrete typing units of Trypanosoma cruzi. Parasitol Res 2024; 123:181. [PMID: 38602595 PMCID: PMC11008065 DOI: 10.1007/s00436-024-08183-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/08/2024] [Indexed: 04/12/2024]
Abstract
Chagas disease (CD), caused by the complex life cycle parasite Trypanosoma cruzi, is a global health concern and impacts millions globally. T. cruzi's genetic variability is categorized into discrete typing units (DTUs). Despite their widespread presence in the Americas, a comprehensive understanding of their impact on CD is lacking. This study aims to analyze life cycle traits across life cycle stages, unraveling DTU dynamics. Metacyclogenesis curves were generated, inducing nutritional stress in epimastigotes of five DTUs (TcI (MG), TcI (DA), TcII(Y), TcIII, TcIV, and TcVI), resulting in metacyclic trypomastigotes. Infection dynamics in Vero cells from various DTUs were evaluated, exploring factors like amastigotes per cell, cell-derived trypomastigotes, and infection percentage. Statistical analyses, including ANOVA tests, identified significant differences. Varying onset times for metacyclogenesis converged on the 7th day. TcI (MG) exhibited the highest metacyclogenesis potential. TcI (DA) stood out, infecting 80% of cells within 24 h. TcI demonstrated the highest potential in both metacyclogenesis and infection among the strains assessed. Intra-DTU diversity was evident among TcI strains, contributing to a comprehensive understanding of Trypanosoma cruzi dynamics and genetic diversity.
Collapse
Affiliation(s)
- Tatiana M Cáceres
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Lissa Cruz-Saavedra
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Luz Helena Patiño
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia.
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
21
|
Durães-Oliveira J, Palma-Marques J, Moreno C, Rodrigues A, Monteiro M, Alexandre-Pires G, da Fonseca IP, Santos-Gomes G. Chagas Disease: A Silent Threat for Dogs and Humans. Int J Mol Sci 2024; 25:3840. [PMID: 38612650 PMCID: PMC11011309 DOI: 10.3390/ijms25073840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/15/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Chagas disease (CD) is a vector-borne Neglected Zoonotic Disease (NZD) caused by a flagellate protozoan, Trypanosoma cruzi, that affects various mammalian species across America, including humans and domestic animals. However, due to an increase in population movements and new routes of transmission, T. cruzi infection is presently considered a worldwide health concern, no longer restricted to endemic countries. Dogs play a major role in the domestic cycle by acting very efficiently as reservoirs and allowing the perpetuation of parasite transmission in endemic areas. Despite the significant progress made in recent years, still there is no vaccine against human and animal disease, there are few drugs available for the treatment of human CD, and there is no standard protocol for the treatment of canine CD. In this review, we highlight human and canine Chagas Disease in its different dimensions and interconnections. Dogs, which are considered to be the most important peridomestic reservoir and sentinel for the transmission of T. cruzi infection in a community, develop CD that is clinically similar to human CD. Therefore, an integrative approach, based on the One Health concept, bringing together the advances in genomics, immunology, and epidemiology can lead to the effective development of vaccines, new treatments, and innovative control strategies to tackle CD.
Collapse
Affiliation(s)
- João Durães-Oliveira
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008 Lisbon, Portugal; (J.D.-O.); (G.S.-G.)
| | - Joana Palma-Marques
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008 Lisbon, Portugal; (J.D.-O.); (G.S.-G.)
| | - Cláudia Moreno
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008 Lisbon, Portugal; (J.D.-O.); (G.S.-G.)
| | - Armanda Rodrigues
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008 Lisbon, Portugal; (J.D.-O.); (G.S.-G.)
| | - Marta Monteiro
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008 Lisbon, Portugal; (J.D.-O.); (G.S.-G.)
- Centre for Interdisciplinary Research in Animal Health, CIISA, Faculty of Veterinary Medicine, FMV, University of Lisbon, ULisboa, 1649-004 Lisbon, Portugal; (G.A.-P.); (I.P.d.F.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Graça Alexandre-Pires
- Centre for Interdisciplinary Research in Animal Health, CIISA, Faculty of Veterinary Medicine, FMV, University of Lisbon, ULisboa, 1649-004 Lisbon, Portugal; (G.A.-P.); (I.P.d.F.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Isabel Pereira da Fonseca
- Centre for Interdisciplinary Research in Animal Health, CIISA, Faculty of Veterinary Medicine, FMV, University of Lisbon, ULisboa, 1649-004 Lisbon, Portugal; (G.A.-P.); (I.P.d.F.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Gabriela Santos-Gomes
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008 Lisbon, Portugal; (J.D.-O.); (G.S.-G.)
| |
Collapse
|
22
|
Gómez-Palacio A, Cruz-Saavedra L, Van den Broeck F, Geerts M, Pita S, Vallejo GA, Carranza JC, Ramírez JD. High-throughput analysis of the Trypanosoma cruzi minicirculome (mcDNA) unveils structural variation and functional diversity. Sci Rep 2024; 14:5578. [PMID: 38448494 PMCID: PMC10917808 DOI: 10.1038/s41598-024-56076-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 03/01/2024] [Indexed: 03/08/2024] Open
Abstract
Trypanosoma cruzi causes Chagas disease and has a unique extranuclear genome enclosed in a structure called the kinetoplast, which contains circular genomes known as maxi- and minicircles. While the structure and function of maxicircles are well-understood, many aspects of minicircles remain to be discovered. Here, we performed a high-throughput analysis of the minicirculome (mcDNA) in 50 clones isolated from Colombia's diverse T. cruzi I populations. Results indicate that mcDNA comprises four diverse subpopulations with different structures, lengths, and numbers of interspersed semi-conserved (previously termed ultra-conserved regions mHCV) and hypervariable (mHVPs) regions. Analysis of mcDNA ancestry and inter-clone differentiation indicates the interbreeding of minicircle sequence classes is placed along diverse strains and hosts. These results support evidence of the multiclonal dynamics and random bi-parental segregation. Finally, we disclosed the guide RNA repertoire encoded by mcDNA at a clonal scale, and several attributes of its abundance and function are discussed.
Collapse
Affiliation(s)
- Andrés Gómez-Palacio
- Laboratorio de Investigación en Genética Evolutiva, Universidad Pedagógica y Tecnológica de Colombia, Boyacá, Colombia
| | - Lissa Cruz-Saavedra
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Frederik Van den Broeck
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, 3000, Leuven, Belgium
- Department of Biomedical Sciences, Institute of Tropical Medicine, 2000, Antwerp, Belgium
| | - Manon Geerts
- Fish Eco-Evo-Devo and Conservation, KU Leuven, 3000, Leuven, Belgium
- Directorate Taxonomy and Phylogeny, Royal Belgian Institute for Natural Sciences, 1000, Brussels, Belgium
| | - Sebastián Pita
- Sección Genética Evolutiva, Facultad de Ciencias, Universidad de La República, Montevideo, Uruguay
| | - Gustavo A Vallejo
- Laboratorio de Investigación en Parasitología Tropical, Facultad de Ciencias, Universidad del Tolima, Ibagué, Colombia
| | - Julio C Carranza
- Laboratorio de Investigación en Parasitología Tropical, Facultad de Ciencias, Universidad del Tolima, Ibagué, Colombia
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia.
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York City, NY, 10029, USA.
| |
Collapse
|
23
|
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
|
24
|
Reifler KA, Wheelock A, Hall SM, Salazar A, Hassan S, Bostrom JA, Barnett ED, Carrion M, Hochberg NS, Hamer DH, Gopal DM, Bourque D. Chagas cardiomyopathy in Boston, Massachusetts: Identifying disease and improving management after community and hospital-based screening. PLoS Negl Trop Dis 2024; 18:e0011913. [PMID: 38241361 PMCID: PMC10830043 DOI: 10.1371/journal.pntd.0011913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/31/2024] [Accepted: 01/11/2024] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND Limited data exist regarding cardiac manifestations of Chagas disease in migrants living in non-endemic regions. METHODS A retrospective cohort analysis of 109 patients with Chagas disease seen at Boston Medical Center (BMC) between January 2016 and January 2023 was performed. Patients were identified by screening and testing migrants from endemic regions at a community health center and BMC. Demographic, laboratory, and cardiac evaluation data were collected. RESULTS Mean age of the 109 patients was 43 years (range 19-76); 61% were female. 79% (86/109) were diagnosed with Chagas disease via screening and 21% (23/109) were tested given symptoms or electrocardiogram abnormalities. Common symptoms included palpitations (25%, 27/109) and chest pain (17%, 18/109); 52% (57/109) were asymptomatic. Right bundle branch block (19%, 19/102), T-wave changes (18%, 18/102), and left anterior fascicular block (11%, 11/102) were the most common electrocardiogram abnormalities; 51% (52/102) had normal electrocardiograms. Cardiomyopathy stage was ascertained in 94 of 109 patients: 51% (48/94) were indeterminate stage A and 49% (46/94) had cardiac structural disease (stages B1-D). Clinical findings that required clinical intervention or change in management were found in 23% (25/109), and included cardiomyopathy, apical hypokinesis/aneurysm, stroke, atrial or ventricular arrhythmias, and apical thrombus. CONCLUSIONS These data show high rates of cardiac complications in a cohort of migrants living with Chagas disease in a non-endemic setting. We demonstrate that Chagas disease diagnosis prompts cardiac evaluation which often identifies actionable cardiac disease and provides opportunities for prevention and treatment.
Collapse
Affiliation(s)
- Katherine A. Reifler
- Section of Infectious Disease, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| | - Alyse Wheelock
- Section of Preventative Medicine and Epidemiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| | - Samantha M. Hall
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Alejandra Salazar
- Section of Infectious Disease, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| | - Shahzad Hassan
- Department of Internal Medicine, Boston University Medical Center, Boston, Massachusetts, United States of America
| | - John A. Bostrom
- Cardiovascular Division, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| | - Elizabeth D. Barnett
- Section of Pediatric Infectious Disease, Department of Pediatrics, Boston Medical Center and Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| | - Malwina Carrion
- Boston University College of Health and Rehabilitation Sciences: Sargent College, Massachusetts, United States of America
| | - Natasha S. Hochberg
- Section of Infectious Disease, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| | - Davidson H. Hamer
- Section of Infectious Disease, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
- Department of Global Health, Boston University School of Public Health, Boston, Massachusetts, United States of America
- Center for Emerging Infectious Disease Policy & Research, Boston University, Boston, Massachusetts, United States of America
| | - Deepa M. Gopal
- Cardiovascular Division, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| | - Daniel Bourque
- Section of Infectious Disease, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
25
|
Zingales B, Macedo AM. Fifteen Years after the Definition of Trypanosoma cruzi DTUs: What Have We Learned? Life (Basel) 2023; 13:2339. [PMID: 38137940 PMCID: PMC10744745 DOI: 10.3390/life13122339] [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: 10/30/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Trypanosoma cruzi, the protozoan causative of Chagas disease (ChD), exhibits striking genetic and phenotypic intraspecific diversity, along with ecoepidemiological complexity. Human-pathogen interactions lead to distinct clinical presentations of ChD. In 2009, an international consensus classified T. cruzi strains into six discrete typing units (DTUs), TcI to TcVI, later including TcBat, and proposed reproducible genotyping schemes for DTU identification. This article aims to review the impact of classifying T. cruzi strains into DTUs on our understanding of biological, ecoepidemiological, and pathogenic aspects of T. cruzi. We will explore the likely origin of DTUs and the intrinsic characteristics of each group of strains concerning genome organization, genomics, and susceptibility to drugs used in ChD treatment. We will also provide an overview of the association of DTUs with mammalian reservoirs, and summarize the geographic distribution, and the clinical implications, of prevalent specific DTUs in ChD patients. Throughout this review, we will emphasize the crucial roles of both parasite and human genetics in defining ChD pathogenesis and chemotherapy outcome.
Collapse
Affiliation(s)
- Bianca Zingales
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo 05508-900, São Paulo, Brazil
| | - Andréa M. Macedo
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil;
| |
Collapse
|
26
|
García-López C, Santos-Hernández NG, Gutiérrez-Jiménez J, Vidal-López DG, Moreno-Rodríguez A, Gutiérrez-Cabrera AE, De Fuentes-Vicente JA. Identification of Discrete Typing Units of Trypanosoma cruzi Isolated from Domestic Environments in Southeastern Mexico. Vector Borne Zoonotic Dis 2023. [PMID: 38052044 DOI: 10.1089/vbz.2023.0075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023] Open
Abstract
Background: The Trypanosoma cruzi parasite is the causal agent of Chagas disease, recognized by the World Health Organization as a neglected tropical disease. Currently there are seven discrete typing units (DTUs) of T. cruzi distributed in America, but there are still gaps about its distribution in some endemic regions. Materials and Methods: Seventeen units isolated from Chiapas and Oaxaca in Mexico were identified by amplification of the C-5 sterol desaturase gene. Results: Three DTUs of T. cruzi, TcI (6), TcII (10), and TcIV (1) were detected by comparing polymorphic sites in specific regions. Conclusions: New DTUs are reported for both states, where TcII was the most common DTU. The genetic characterization of the isolates can help to understand the epidemiology of Chagas disease.
Collapse
Affiliation(s)
- Carlos García-López
- Instituto de Ciencias Biológicas, Universidad de Ciencias y Artes de Chiapas, Tuxtla Gutierrez, México
| | - Nancy G Santos-Hernández
- Instituto de Ciencias Biológicas, Universidad de Ciencias y Artes de Chiapas, Tuxtla Gutierrez, México
| | - Javier Gutiérrez-Jiménez
- Instituto de Ciencias Biológicas, Universidad de Ciencias y Artes de Chiapas, Tuxtla Gutierrez, México
| | - Dolores G Vidal-López
- Instituto de Ciencias Biológicas, Universidad de Ciencias y Artes de Chiapas, Tuxtla Gutierrez, México
| | | | - Ana E Gutiérrez-Cabrera
- Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, México
| | - José A De Fuentes-Vicente
- Instituto de Ciencias Biológicas, Universidad de Ciencias y Artes de Chiapas, Tuxtla Gutierrez, México
| |
Collapse
|
27
|
Duque C, So J, Castro-Sesquen YE, DeToy K, Gutierrez Guarnizo SA, Jahanbakhsh F, Machaca EM, Miranda-Schaeubinger M, Chakravarti I, Cooper V, Schmidt ME, Adamo L, Marcus R, Talaat KR, Gilman RH, Mugnier MR. Immunologic changes are detectable in the peripheral blood transcriptome of clinically asymptomatic Chagas cardiomyopathy patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.03.560680. [PMID: 37873108 PMCID: PMC10592925 DOI: 10.1101/2023.10.03.560680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Chagas disease, caused by the protozoan parasite Trypanosoma cruzi, is a neglected parasitic disease that affects approximately 6 million individuals worldwide. Of those infected, 20-30% will go on to develop chronic Chagas cardiomyopathy (CCC), and ultimately many of these individuals will progress to advanced heart failure. The mechanism by which this progression occurs is poorly understood, as few studies have focused on early CCC. In this study, we sought to understand the physiologic changes associated with T. cruzi infection and the development of CCC. We analyzed gene expression in the peripheral blood of asymptomatic Chagas patients with early structural heart disease, Chagas patients without any signs or symptoms of disease, and Chagas-negative patients with and without early structural heart disease. Our analysis shows that early CCC was associated with a downregulation of various peripheral immune response genes, with gene expression changes suggestive of reduced antigen presentation and T cell activation. Notably, these genes and processes were distinct from those of early cardiomyopathy in Chagas-negative patients, suggesting that the processes mediating CCC may be unique from those mediating progression to other cardiomyopathies. This work highlights the importance of the immune response in early CCC, providing insight into the early pathogenesis of this disease. The changes we have identified may serve as biomarkers of progression and could inform strategies for the treatment of CCC in its early stages, before significant cardiac damage has occurred.
Collapse
Affiliation(s)
- Carolina Duque
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Jaime So
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Yagahira E Castro-Sesquen
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Kelly DeToy
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | | | - Fatemeh Jahanbakhsh
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Edith Malaga Machaca
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Monica Miranda-Schaeubinger
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Indira Chakravarti
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Virginia Cooper
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Mary E Schmidt
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Luigi Adamo
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rachel Marcus
- MedStar Washington Hospital Center, Washington, D.C., USA
| | - Kawsar R Talaat
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Robert H Gilman
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Monica R Mugnier
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
28
|
Fesser A, Beilstein S, Kaiser M, Schmidt RS, Mäser P. Trypanosoma cruzi STIB980: A TcI Strain for Drug Discovery and Reverse Genetics. Pathogens 2023; 12:1217. [PMID: 37887733 PMCID: PMC10610277 DOI: 10.3390/pathogens12101217] [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: 08/29/2023] [Revised: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 10/28/2023] Open
Abstract
Since the first published genome sequence of Trypanosoma cruzi in 2005, there have been tremendous technological advances in genomics, reverse genetics, and assay development for this elusive pathogen. However, there is still an unmet need for new and better drugs to treat Chagas disease. Here, we introduce a T. cruzi assay strain that is useful for drug research and basic studies of host-pathogen interactions. T. cruzi STIB980 is a strain of discrete typing unit TcI that grows well in culture as axenic epimastigotes or intracellular amastigotes. We evaluated the optimal parameters for genetic transfection and constructed derivatives of T. cruzi STIB980 that express reporter genes for fluorescence- or bioluminescence-based drug efficacy testing, as well as a Cas9-expressing line for CRISPR/Cas9-mediated gene editing. The genome of T. cruzi STIB980 was sequenced by combining short-read Illumina with long-read Oxford Nanopore technologies. The latter served as the primary assembly and the former to correct mistakes. This resulted in a high-quality nuclear haplotype assembly of 28 Mb in 400 contigs, containing 10,043 open-reading frames with a median length of 1077 bp. We believe that T. cruzi STIB980 is a useful addition to the antichagasic toolbox and propose that it can serve as a DTU TcI reference strain for drug efficacy testing.
Collapse
Affiliation(s)
- Anna Fesser
- Swiss Tropical and Public Health Institute, Department Medical Parasitology and Infection Biology, 4123 Allschwil, Switzerland
- University of Basel, 4001 Basel, Switzerland
| | - Sabina Beilstein
- Swiss Tropical and Public Health Institute, Department Medical Parasitology and Infection Biology, 4123 Allschwil, Switzerland
- University of Basel, 4001 Basel, Switzerland
| | - Marcel Kaiser
- Swiss Tropical and Public Health Institute, Department Medical Parasitology and Infection Biology, 4123 Allschwil, Switzerland
- University of Basel, 4001 Basel, Switzerland
| | - Remo S Schmidt
- Swiss Tropical and Public Health Institute, Department Medical Parasitology and Infection Biology, 4123 Allschwil, Switzerland
- University of Basel, 4001 Basel, Switzerland
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute, Department Medical Parasitology and Infection Biology, 4123 Allschwil, Switzerland
- University of Basel, 4001 Basel, Switzerland
| |
Collapse
|
29
|
Gomes DC, Medeiros TS, Alves Pereira EL, da Silva JFO, de Freitas Oliveira JW, Fernandes-Pedrosa MDF, de Sousa da Silva M, da Silva-Júnior AA. From Benznidazole to New Drugs: Nanotechnology Contribution in Chagas Disease. Int J Mol Sci 2023; 24:13778. [PMID: 37762080 PMCID: PMC10530915 DOI: 10.3390/ijms241813778] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/31/2023] [Accepted: 08/05/2023] [Indexed: 09/29/2023] Open
Abstract
Chagas disease is a neglected tropical disease caused by the protozoan Trypanosoma cruzi. Benznidazole and nifurtimox are the two approved drugs for their treatment, but both drugs present side effects and efficacy problems, especially in the chronic phase of this disease. Therefore, new molecules have been tested with promising results aiming for strategic targeting action against T. cruzi. Several studies involve in vitro screening, but a considerable number of in vivo studies describe drug bioavailability increment, drug stability, toxicity assessment, and mainly the efficacy of new drugs and formulations. In this context, new drug delivery systems, such as nanotechnology systems, have been developed for these purposes. Some nanocarriers are able to interact with the immune system of the vertebrate host, modulating the immune response to the elimination of pathogenic microorganisms. In this overview of nanotechnology-based delivery strategies for established and new antichagasic agents, different strategies, and limitations of a wide class of nanocarriers are explored, as new perspectives in the treatment and monitoring of Chagas disease.
Collapse
Affiliation(s)
- Daniele Cavalcante Gomes
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte-UFRN, Natal 59012-570, Brazil; (D.C.G.); (T.S.M.); (E.L.A.P.); (J.F.O.d.S.); (M.d.F.F.-P.)
| | - Thayse Silva Medeiros
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte-UFRN, Natal 59012-570, Brazil; (D.C.G.); (T.S.M.); (E.L.A.P.); (J.F.O.d.S.); (M.d.F.F.-P.)
| | - Eron Lincoln Alves Pereira
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte-UFRN, Natal 59012-570, Brazil; (D.C.G.); (T.S.M.); (E.L.A.P.); (J.F.O.d.S.); (M.d.F.F.-P.)
| | - João Felipe Oliveira da Silva
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte-UFRN, Natal 59012-570, Brazil; (D.C.G.); (T.S.M.); (E.L.A.P.); (J.F.O.d.S.); (M.d.F.F.-P.)
| | - Johny W. de Freitas Oliveira
- Immunoparasitology Laboratory, Department of Clinical and Toxicological Analysis, Centre of Health Sciences, Federal University of Rio Grande do Norte-UFRN, Natal 59012-570, Brazil; (J.W.d.F.O.); (M.d.S.d.S.)
| | - Matheus de Freitas Fernandes-Pedrosa
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte-UFRN, Natal 59012-570, Brazil; (D.C.G.); (T.S.M.); (E.L.A.P.); (J.F.O.d.S.); (M.d.F.F.-P.)
| | - Marcelo de Sousa da Silva
- Immunoparasitology Laboratory, Department of Clinical and Toxicological Analysis, Centre of Health Sciences, Federal University of Rio Grande do Norte-UFRN, Natal 59012-570, Brazil; (J.W.d.F.O.); (M.d.S.d.S.)
| | - Arnóbio Antônio da Silva-Júnior
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte-UFRN, Natal 59012-570, Brazil; (D.C.G.); (T.S.M.); (E.L.A.P.); (J.F.O.d.S.); (M.d.F.F.-P.)
| |
Collapse
|
30
|
Romer G, Bracco LA, Ricci AD, Balouz V, Berná L, Villar JC, Ramsey JM, Nolan MS, Torrico F, Kesper N, Altcheh J, Robello C, Buscaglia CA, Agüero F. Deep serological profiling of the Trypanosoma cruzi TSSA antigen reveals different epitopes and modes of recognition by Chagas disease patients. PLoS Negl Trop Dis 2023; 17:e0011542. [PMID: 37556493 PMCID: PMC10441789 DOI: 10.1371/journal.pntd.0011542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 08/21/2023] [Accepted: 07/18/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Trypanosoma cruzi, the agent of Chagas disease, displays a highly structured population, with multiple strains that can be grouped into 6-7 evolutionary lineages showing variable eco-epidemiological traits and likely also distinct disease-associated features. Previous works have shown that antibody responses to 'isoforms' of the polymorphic parasite antigen TSSA enable robust and sensitive identification of the infecting strain with near lineage-level resolution. To optimize the serotyping performance of this molecule, we herein used a combination of immunosignaturing approaches based on peptide microarrays and serum samples from Chagas disease patients to establish a deep linear B-cell epitope profiling of TSSA. METHODS/PRINCIPLE FINDINGS Our assays revealed variations in the seroprevalence of TSSA isoforms among Chagas disease populations from different settings, hence strongly supporting the differential distribution of parasite lineages in domestic cycles across the Americas. Alanine scanning mutagenesis and the use of peptides of different lengths allowed us to identify key residues involved in antibody pairing and the presence of three discrete B-cell linear epitopes in TSSAII, the isoform with highest seroprevalence in human infections. Comprehensive screening of parasite genomic repositories led to the discovery of 9 novel T. cruzi TSSA variants and one TSSA sequence from the phylogenetically related bat parasite T. cruzi marinkellei. Further residue permutation analyses enabled the identification of diagnostically relevant or non-relevant substitutions among TSSA natural polymorphisms. Interestingly, T. cruzi marinkellei TSSA displayed specific serorecognition by one chronic Chagas disease patient from Colombia, which warrant further investigations on the diagnostic impact of such atypical TSSA. CONCLUSIONS/SIGNIFICANCE Overall, our findings shed new light into TSSA evolution, epitope landscape and modes of recognition by Chagas disease patients; and have practical implications for the design and/or evaluation of T. cruzi serotyping strategies.
Collapse
Affiliation(s)
- Guadalupe Romer
- Instituto de Investigaciones Biotecnológicas (IIB)–Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), B1650HMP, San Martín, Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad de San Martín (UNSAM), San Martín, Buenos Aires, Argentina
| | - Leonel A. Bracco
- Instituto de Investigaciones Biotecnológicas (IIB)–Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), B1650HMP, San Martín, Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad de San Martín (UNSAM), San Martín, Buenos Aires, Argentina
| | - Alejandro D. Ricci
- Instituto de Investigaciones Biotecnológicas (IIB)–Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), B1650HMP, San Martín, Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad de San Martín (UNSAM), San Martín, Buenos Aires, Argentina
| | - Virginia Balouz
- Instituto de Investigaciones Biotecnológicas (IIB)–Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), B1650HMP, San Martín, Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad de San Martín (UNSAM), San Martín, Buenos Aires, Argentina
| | - Luisa Berná
- Laboratorio de Interacciones Hospedero-Patógeno, Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Sección Biomatemática-Unidad de Genómica Evolutiva, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Juan C. Villar
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga y Fundación Cardioinfantil—Instituto de Cardiología, Colombia
| | - Janine M. Ramsey
- Centro Regional de Investigación en Salud Pública, Instituto Nacional de Salud Pública, Tapachula, México
| | - Melissa S. Nolan
- Laboratory of Vector-borne and Zoonotic Diseases, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, United States of America
| | | | - Norival Kesper
- LIM-49, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São 33 Paulo, São Paulo, Brazil
| | - Jaime Altcheh
- Hospital de Niños “Ricardo Gutierrez”, Ciudad Autónoma de Buenos Aires, Argentina
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas (IMIPP)–GCBA-CONICET, Buenos Aires, Argentina
| | - Carlos Robello
- Laboratorio de Interacciones Hospedero-Patógeno, Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Carlos A. Buscaglia
- Instituto de Investigaciones Biotecnológicas (IIB)–Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), B1650HMP, San Martín, Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad de San Martín (UNSAM), San Martín, Buenos Aires, Argentina
| | - Fernán Agüero
- Instituto de Investigaciones Biotecnológicas (IIB)–Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), B1650HMP, San Martín, Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad de San Martín (UNSAM), San Martín, Buenos Aires, Argentina
| |
Collapse
|
31
|
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
|
32
|
Arce-Fonseca M, Gutiérrez-Ocejo RA, Rosales-Encina JL, Aranda-Fraustro A, Cabrera-Mata JJ, Rodríguez-Morales O. Nitazoxanide: A Drug Repositioning Compound with Potential Use in Chagas Disease in a Murine Model. Pharmaceuticals (Basel) 2023; 16:826. [PMID: 37375773 DOI: 10.3390/ph16060826] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 05/30/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Chagas disease (ChD), caused by Trypanosoma cruzi, is the most serious parasitosis in the western hemisphere. Benznidazole and nifurtimox, the only two trypanocidal drugs, are expensive, difficult to obtain, and have severe side effects. Nitazoxanide has shown to be effective against protozoa, bacteria, and viruses. This study aimed to evaluate the nitazoxanide efficacy against the Mexican T. cruzi Ninoa strain in mice. Infected animals were orally treated for 30 days with nitazoxanide (100 mg/kg) or benznidazole (10 mg/kg). The clinical, immunological, and histopathological conditions of the mice were evaluated. Nitazoxanide- or benznidazole-treated mice had longer survival and less parasitemia than those without treatment. Antibody production in the nitazoxanide-treated mice was of the IgG1-type and not of the IgG2-type as in the benznidazole-treated mice. Nitazoxanide-treated mice had significantly high IFN-γ levels compared to the other infected groups. Serious histological damage could be prevented with nitazoxanide treatment compared to without treatment. In conclusion, nitazoxanide decreased parasitemia levels, indirectly induced the production of IgG antibodies, and partially prevented histopathological damage; however, it did not show therapeutic superiority compared to benznidazole in any of the evaluated aspects. Therefore, the repositioning of nitazoxanide as an alternative treatment against ChD could be considered, since it did not trigger adverse effects that worsened the pathological condition of the infected mice.
Collapse
Affiliation(s)
- Minerva Arce-Fonseca
- Laboratory of Molecular Immunology and Proteomics, Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Rodolfo Andrés Gutiérrez-Ocejo
- Laboratory of Molecular Immunology and Proteomics, Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - José Luis Rosales-Encina
- Department of Infectomics and Molecular Pathogenesis, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Insituto Politécnico Nacional, Av. Instituto Politécnico Nacional No. 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico
| | - Alberto Aranda-Fraustro
- Department of Pathology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Juan José Cabrera-Mata
- Laboratory of Molecular Immunology and Proteomics, Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Olivia Rodríguez-Morales
- Laboratory of Molecular Immunology and Proteomics, Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| |
Collapse
|
33
|
Pagotti MC, Dias HJ, Candido ACBB, Oliveira TAS, Borges A, Oliveira ND, Lopes CD, Orenha RP, Parreira RLT, Crotti AEM, Magalhães LG. Exploring Synthetic Dihydrobenzofuran and Benzofuran Neolignans as Antiprotozoal Agents against Trypanosoma cruzi. Pharmaceutics 2023; 15:pharmaceutics15030754. [PMID: 36986617 PMCID: PMC10052957 DOI: 10.3390/pharmaceutics15030754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/18/2023] [Accepted: 02/20/2023] [Indexed: 03/30/2023] Open
Abstract
Chagas disease is a neglected tropical disease that affects more than 8 million people. Although there are therapies against this disease, the search for new drugs is important because the current treatments show limited effectiveness and high toxicity. In this work, eighteen dihydrobenzofuran-type neolignans (DBNs) and two benzofuran-type neolignans (BNs) were synthesized and evaluated against amastigote forms of two Trypanosoma cruzi strains. The in vitro cytotoxicity and hemolytic activity of the most active compounds were also evaluated and their relationships with T. cruzi tubulin DBNs were investigated by an in silico approach. Four DBNs demonstrated activity against the T. cruzi Tulahuen lac-Z strain (IC50 from 7.96 to 21.12 µM), and DBN 1 exhibited the highest activity against the amastigote forms of the T. cruzi Y strain (IC50 3.26 μM). Compounds 1-4 showed CC50 values higher than antitrypanosomal activities, except for DBN 3. All DBNs with antitrypanosomal activity demonstrated CH50 higher than 100 µM. The in silico results indicated that DBNs 1, 2, and 4 are capable of destabilizing the dynamics of the tubulin-microtubule from the vinca site. These compounds displayed promising in vitro activity against T. cruzi, especially compound 1, and can be considered molecular prototypes for the development of new antiparasitic drugs.
Collapse
Affiliation(s)
- Mariana C Pagotti
- Research Group on Natural Products, Center for Research in Sciences and Technology, University of Franca, Franca 14404-600, SP, Brazil
| | - Herbert J Dias
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters, University of São Paulo, Ribeirão Preto 14040-901, SP, Brazil
- Goiano Federal Institute of Education, Science, and Technology, Campus Urutaí, Urutaí 75790-000, GO, Brazil
| | - Ana Carolina B B Candido
- Research Group on Natural Products, Center for Research in Sciences and Technology, University of Franca, Franca 14404-600, SP, Brazil
| | - Thaís A S Oliveira
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters, University of São Paulo, Ribeirão Preto 14040-901, SP, Brazil
| | - Alexandre Borges
- Research Group on Natural Products, Center for Research in Sciences and Technology, University of Franca, Franca 14404-600, SP, Brazil
- Faculty of Medicine, University Center of Santa Fe do Sul, Santa Fé do Sul 15775-000, SP, Brazil
| | - Nicoli D Oliveira
- Animal Science Post Graduation, University of Franca, Franca 14404-600, SP, Brazil
| | - Carla D Lopes
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Renato P Orenha
- Research Group on Natural Products, Center for Research in Sciences and Technology, University of Franca, Franca 14404-600, SP, Brazil
| | - Renato L T Parreira
- Research Group on Natural Products, Center for Research in Sciences and Technology, University of Franca, Franca 14404-600, SP, Brazil
| | - Antônio E M Crotti
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters, University of São Paulo, Ribeirão Preto 14040-901, SP, Brazil
| | - Lizandra G Magalhães
- Research Group on Natural Products, Center for Research in Sciences and Technology, University of Franca, Franca 14404-600, SP, Brazil
- Animal Science Post Graduation, University of Franca, Franca 14404-600, SP, Brazil
| |
Collapse
|
34
|
Velásquez-Ortiz N, Hernández C, Cantillo-Barraza O, Ballesteros N, Cruz-Saavedra L, Herrera G, Buitrago LS, Soto H, Medina M, Palacio J, González MS, Cuervo A, Vallejo G, Zuleta Dueñas L, Urbano P, Muñoz M, Ramírez JD. Trypanosoma cruzi Parasite Burdens of Several Triatomine Species in Colombia. Trop Med Infect Dis 2022; 7:tropicalmed7120445. [PMID: 36548700 PMCID: PMC9782637 DOI: 10.3390/tropicalmed7120445] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Trypanosoma cruzi, the causal agent of Chagas disease, is mainly transmitted by insects of the Triatominae subfamily. In Colombia, there are 26 triatomine species, and 16 of them are naturally infected with the parasite. The parasite loads of naturally infected vectors can be significant in targeting specific species that can affect the epidemiology of the disease. Studying their ecology and behavior is vital to understand their role in T. cruzi transmission dynamics. We evaluated the parasite loads of 182 field-collected triatomines corresponding to 10 species in 13 departments across Colombia. We standardized a methodology to quantify T. cruzi DNA in these insects. We obtained a LOD (limit of detection) of 3.05 p-eq/mL. The 82% of triatomines we evaluated were positive for T. cruzi infection, with loads ranging from hundreds to millions of equivalent parasites per milliliter. Panstrongylus geniculatus, Rhodnius prolixus, and Triatoma dimidiata were the species with the highest loads of T. cruzi; however, other species whose role as vectors is still unknown were also found with high loads of parasites. Our results suggest the relevance of secondary species for T. cruzi transmission in Colombia. We hope our data can help improve entomological surveillance and vector control programs in the country and the region.
Collapse
Affiliation(s)
- Natalia Velásquez-Ortiz
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Carolina Hernández
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
- Centro de Tecnología en Salud (CETESA), Innovaseq SAS, Bogotá, Colombia
| | - Omar Cantillo-Barraza
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
- Grupo BCEI, Universidad de Antioquia, Medellín, Colombia
| | - Nathalia Ballesteros
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Lissa Cruz-Saavedra
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Giovanny Herrera
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | | | - Hugo Soto
- Laboratorio de Salud Pública del Cesar, Valledupar, Colombia
| | - Manuel Medina
- Programa de Control de ETV, Secretaría de Salud de Boyacá, Tunja, Colombia
| | - Jatney Palacio
- Laboratorio Departamental de Salud Pública del Departamento del Chocó, Quibdó, Colombia
| | | | - Andrés Cuervo
- Secretaría Departamental de Salud de Arauca, Arauca, Colombia
| | - Gustavo Vallejo
- Laboratorio de Investigaciones en Parasitología Tropical, Facultad de Ciencias, Universidad del Tolima, Ibagué, Colombia
| | - Liliana Zuleta Dueñas
- Grupo de Vigilancia en Salud Pública, Secretaría de Salud de Casanare, Yopal, Colombia
| | - Plutarco Urbano
- Grupo de Investigaciones Biológicas de la Orinoquia, Universidad Internacional del Trópico Americano (Unitrópico), Yopal, Colombia
| | - Marina Muñoz
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- Correspondence: or
| |
Collapse
|
35
|
Rashidi S, Mansouri R, Ali-Hassanzadeh M, Muro A, Nguewa P, Manzano-Román R. The Defensive Interactions of Prominent Infectious Protozoan Parasites: The Host's Complement System. Biomolecules 2022; 12:1564. [PMID: 36358913 PMCID: PMC9687244 DOI: 10.3390/biom12111564] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/16/2022] [Accepted: 10/21/2022] [Indexed: 12/30/2023] Open
Abstract
The complement system exerts crucial functions both in innate immune responses and adaptive humoral immunity. This pivotal system plays a major role dealing with pathogen invasions including protozoan parasites. Different pathogens including parasites have developed sophisticated strategies to defend themselves against complement killing. Some of these strategies include the employment, mimicking or inhibition of host's complement regulatory proteins, leading to complement evasion. Therefore, parasites are proven to use the manipulation of the complement system to assist them during infection and persistence. Herein, we attempt to study the interaction´s mechanisms of some prominent infectious protozoan parasites including Plasmodium, Toxoplasma, Trypanosoma, and Leishmania dealing with the complement system. Moreover, several crucial proteins that are expressed, recruited or hijacked by parasites and are involved in the modulation of the host´s complement system are selected and their role for efficient complement killing or lysis evasion is discussed. In addition, parasite's complement regulatory proteins appear as plausible therapeutic and vaccine targets in protozoan parasitic infections. Accordingly, we also suggest some perspectives and insights useful in guiding future investigations.
Collapse
Affiliation(s)
- Sajad Rashidi
- Molecular and Medicine Research Center, Khomein University of Medical Sciences, Khomein 38811, Iran
- Department of Medical Laboratory Sciences, Khomein University of Medical Sciences, Khomein 38811, Iran
| | - Reza Mansouri
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd 8915173143, Iran
| | - Mohammad Ali-Hassanzadeh
- Department of Immunology, School of Medicine, Jiroft University of Medical Sciences, Jiroft 7861615765, Iran
| | - Antonio Muro
- Infectious and Tropical Diseases Group (e-INTRO), Institute of Biomedical Research of Salamanca-Research Center for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Faculty of Pharmacy, University of Salamanca, 37008 Salamanca, Spain
| | - Paul Nguewa
- Department of Microbiology and Parasitology, ISTUN Institute of Tropical Health, IdiSNA (Navarra Institute for Health Research), University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain
| | - Raúl Manzano-Román
- Infectious and Tropical Diseases Group (e-INTRO), Institute of Biomedical Research of Salamanca-Research Center for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Faculty of Pharmacy, University of Salamanca, 37008 Salamanca, Spain
| |
Collapse
|
36
|
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: 1.7] [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
|
37
|
Parasitemia and Differential Tissue Tropism in Mice Infected with Trypanosoma cruzi Isolates Obtained from Meccus phyllosoma in the State of Oaxaca, Mexico. Pathogens 2022; 11:pathogens11101141. [PMID: 36297198 PMCID: PMC9607563 DOI: 10.3390/pathogens11101141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/20/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022] Open
Abstract
Trypanosoma cruzi is a parasite transmitted by the feces of triatomines. Many triatomine species are found in Mexico, and various T. cruzi variants have been isolated from these species, each showing very different virulence and cell tropism. The isolates were obtained from Meccus phyllosoma specimens in three localities in the state of Oaxaca, Mexico: Tehuantitla, Vixhana, and Guichivere. The virulence of each isolate was assessed by quantifying parasitemia, survival, and histopathologic findings. The lineage of each isolate was identified using the mini-exon gene. The expression of the tssa gene during infection was detected in the heart, esophagus, gastrocnemius, and brain. Our results show that the maximum post-infection parasitemia was higher for the Tehuantitla isolate. On genotyping, all isolates were identified as T. cruzi I. The amastigotes in the heart and gastrocnemius were verified for all isolates, but in the brain only for Tehuantitla and Vixhana. The tssa expression allowed us to detect T. cruzi isolates, for Tehuantitla, predominantly in the heart. For Vixhana, a higher tssa expression was detected in gastrocnemius, and for Guichivere, it was higher in the esophagus. Results show that virulence, tropism, and tssa expression can vary, even when the isolates are derived from the same vector species, in the same region, and at similar altitudes.
Collapse
|