Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 18.057
Filtrar
Más filtros

Intervalo de año de publicación
1.
Traffic ; 25(4): e12935, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38629580

RESUMEN

The protozoan parasites Plasmodium falciparum, Leishmania spp. and Trypanosoma cruzi continue to exert a significant toll on the disease landscape of the human population in sub-Saharan Africa and Latin America. Control measures have helped reduce the burden of their respective diseases-malaria, leishmaniasis and Chagas disease-in endemic regions. However, the need for new drugs, innovative vaccination strategies and molecular markers of disease severity and outcomes has emerged because of developing antimicrobial drug resistance, comparatively inadequate or absent vaccines, and a lack of trustworthy markers of morbid outcomes. Extracellular vesicles (EVs) have been widely reported to play a role in the biology and pathogenicity of P. falciparum, Leishmania spp. and T. cruzi ever since they were discovered. EVs are secreted by a yet to be fully understood mechanism in protozoans into the extracellular milieu and carry a cargo of diverse molecules that reflect the originator cell's metabolic state. Although our understanding of the biogenesis and function of EVs continues to deepen, the question of how EVs in P. falciparum, Leishmania spp. and T. cruzi can serve as targets for a translational agenda into clinical and public health interventions is yet to be fully explored. Here, as a consortium of protozoan researchers, we outline a plan for future researchers and pose three questions to direct an EV's translational agenda in P. falciparum, Leishmania spp. and T. cruzi. We opine that in the long term, executing this blueprint will help bridge the current unmet needs of these medically important protozoan diseases in sub-Saharan Africa and Latin America.


Asunto(s)
Enfermedad de Chagas , Vesículas Extracelulares , Leishmania , Parásitos , Trypanosoma cruzi , Animales , Humanos , Enfermedad de Chagas/epidemiología , Enfermedad de Chagas/parasitología
2.
PLoS Pathog ; 20(3): e1012012, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38457443

RESUMEN

Small molecules (less than 1,500 Da) include major biological signals that mediate host-pathogen-microbiome communication. They also include key intermediates of metabolism and critical cellular building blocks. Pathogens present with unique nutritional needs that restrict pathogen colonization or promote tissue damage. In parallel, parts of host metabolism are responsive to immune signaling and regulated by immune cascades. These interactions can trigger both adaptive and maladaptive metabolic changes in the host, with microbiome-derived signals also contributing to disease progression. In turn, targeting pathogen metabolic needs or maladaptive host metabolic changes is an important strategy to develop new treatments for infectious diseases. Trypanosoma cruzi is a single-celled eukaryotic pathogen and the causative agent of Chagas disease, a neglected tropical disease associated with cardiac and intestinal dysfunction. Here, we discuss the role of small molecules during T. cruzi infection in its vector and in the mammalian host. We integrate these findings to build a theoretical interpretation of how maladaptive metabolic changes drive Chagas disease and extrapolate on how these findings can guide drug development.


Asunto(s)
Enfermedad de Chagas , Microbiota , Trypanosoma cruzi , Animales , Humanos , Interacción Gen-Ambiente , Enfermedad de Chagas/metabolismo , Trypanosoma cruzi/metabolismo , Progresión de la Enfermedad , Mamíferos
3.
PLoS Pathog ; 20(4): e1012191, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38683845

RESUMEN

An imbalance between suppressor and effector immune responses may preclude cure in chronic parasitic diseases. In the case of Trypanosoma cruzi infection, specialized regulatory Foxp3+ T (Treg) cells suppress protective type-1 effector responses. Herein, we investigated the kinetics and underlying mechanisms behind the regulation of protective parasite-specific CD8+ T cell immunity during acute T. cruzi infection. Using the DEREG mouse model, we found that Treg cells play a role during the initial stages after T. cruzi infection, restraining the magnitude of CD8+ T cell responses and parasite control. Early Treg cell depletion increased the frequencies of polyfunctional short-lived, effector T cell subsets, without affecting memory precursor cell formation or the expression of activation, exhaustion and functional markers. In addition, Treg cell depletion during early infection minimally affected the antigen-presenting cell response but it boosted CD4+ T cell responses before the development of anti-parasite effector CD8+ T cell immunity. Crucially, the absence of CD39 expression on Treg cells significantly bolstered effector parasite-specific CD8+ T cell responses, preventing increased parasite replication in T. cruzi infected mice adoptively transferred with Treg cells. Our work underscores the crucial role of Treg cells in regulating protective anti-parasite immunity and provides evidence that CD39 expression by Treg cells represents a key immunomodulatory mechanism in this infection model.


Asunto(s)
Antígenos CD , Apirasa , Linfocitos T CD8-positivos , Enfermedad de Chagas , Linfocitos T Reguladores , Trypanosoma cruzi , Animales , Enfermedad de Chagas/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T CD8-positivos/inmunología , Ratones , Trypanosoma cruzi/inmunología , Antígenos CD/inmunología , Antígenos CD/metabolismo , Apirasa/inmunología , Apirasa/metabolismo , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad
4.
Circ Res ; 134(10): 1379-1397, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38723031

RESUMEN

Chagas cardiomyopathy caused by infection with the intracellular parasite Trypanosoma cruzi is the most common and severe expression of human Chagas disease. Heart failure, systemic and pulmonary thromboembolism, arrhythmia, and sudden cardiac death are the principal clinical manifestations of Chagas cardiomyopathy. Ventricular arrhythmias contribute significantly to morbidity and mortality and are the major cause of sudden cardiac death. Significant gaps still exist in the understanding of the pathogenesis mechanisms underlying the arrhythmogenic manifestations of Chagas cardiomyopathy. This article will review the data from experimental studies and translate those findings to draw hypotheses about clinical observations. Human- and animal-based studies at molecular, cellular, tissue, and organ levels suggest 5 main pillars of remodeling caused by the interaction of host and parasite: immunologic, electrical, autonomic, microvascular, and contractile. Integrating these 5 remodeling processes will bring insights into the current knowledge in the field, highlighting some key features for future management of this arrhythmogenic disease.


Asunto(s)
Arritmias Cardíacas , Cardiomiopatía Chagásica , Humanos , Animales , Arritmias Cardíacas/etiología , Arritmias Cardíacas/parasitología , Arritmias Cardíacas/fisiopatología , Cardiomiopatía Chagásica/parasitología , Trypanosoma cruzi/patogenicidad , Enfermedad de Chagas/complicaciones , Enfermedad de Chagas/parasitología , Enfermedad de Chagas/inmunología
5.
J Immunol ; 212(4): 617-631, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38197653

RESUMEN

Chagas disease by Trypanosoma cruzi infection is a major public health issue. The available therapeutic agents have limited efficacy and significant side effects. A reliable vaccine would reduce the threat of T. cruzi infections and prevent Chagas disease. Understanding the immune response to this infection would improve vaccine design. We previously demonstrated that adoptively transferred NK cells from mice immunized with highly attenuated T. cruzi, GFP-DDDHA strain, provided potent protection in naive recipients against secondary lethal challenge with various wild-type (WT) strains. To understand the importance of NK cells in protecting mice against T. cruzi infection, we performed an in-depth characterization of NK cell phenotype, responses, and memory-like traits during acute infections due to GFP-DDDHA and WT strains and in immunized mice during a recall response to a WT lethal challenge. NK cells robustly expanded and became more mature and cytolytic during the GFP-DDDHA strain immunization. NK cells in immunized mice responded more robustly after WT lethal challenge than during an acute primary WT infection. In addition, protection by immunization with the GFP-DDDHA strain is significantly weakened in NK cell-deficient mice and did not prevent parasitemia from WT lethal challenge, indicating that NK cells with memory-like traits were a critical component for early control of WT lethal challenge. Prior T. cruzi vaccine development studies have not included studies of this rapid NK response. These findings provide insights into overcoming existing challenges in developing a safe and effective vaccine to prevent this infection.


Asunto(s)
Enfermedad de Chagas , Vacunas Antiprotozoos , Trypanosoma cruzi , Animales , Ratones , Enfermedad de Chagas/prevención & control , Inmunización , Células Asesinas Naturales
6.
Clin Microbiol Rev ; 37(2): e0009923, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38546225

RESUMEN

SUMMARYAs Chagas disease remains prevalent in the Americas, it is important that healthcare professionals and researchers are aware of the screening, diagnosis, monitoring, and treatment recommendations for the populations of patients they care for and study. Management of Trypanosoma cruzi infection in immunocompromised hosts is challenging, particularly because, regardless of antitrypanosomal treatment status, immunocompromised patients with Chagas disease are at risk for T. cruzi reactivation, which can be lethal. Evidence-based practices to prevent and manage T. cruzi reactivation vary depending on the type of immunocompromise. Here, we review available data describing Chagas disease epidemiology, testing, and management practices for various populations of immunocompromised individuals, including people with HIV and patients undergoing solid organ and hematopoietic stem cell transplantation.


Asunto(s)
Enfermedad de Chagas , Huésped Inmunocomprometido , Humanos , Enfermedad de Chagas/diagnóstico , Enfermedad de Chagas/epidemiología , Enfermedad de Chagas/inmunología , Enfermedad de Chagas/terapia , Trypanosoma cruzi/inmunología
7.
Lancet ; 403(10422): 203-218, 2024 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-38071985

RESUMEN

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.


Asunto(s)
Enfermedad de Chagas , Humanos , Enfermedad de Chagas/diagnóstico , Enfermedad de Chagas/epidemiología , Enfermedad de Chagas/terapia , Política Pública , Salud Pública , Transmisión Vertical de Enfermedad Infecciosa/prevención & control
8.
PLoS Pathog ; 19(11): e1011627, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37956215

RESUMEN

Benznidazole is the front-line drug used to treat infections with Trypanosoma cruzi, the causative agent of Chagas disease. However, for reasons that are unknown, treatment failures are common. When we examined parasites that survived benznidazole treatment in mice using highly sensitive in vivo and ex vivo bioluminescence imaging, we found that recrudescence is not due to persistence of parasites in a specific organ or tissue that preferentially protects them from drug activity. Surviving parasites are widely distributed and located in host cells where the vast majority contained only one or two amastigotes. Therefore, infection relapse does not arise from a small number of intact large nests. Rather, persisters are either survivors of intracellular populations where co-located parasites have been killed, or amastigotes in single/low-level infected cells exist in a state where they are less susceptible to benznidazole. To better assess the nature of parasite persisters, we exposed infected mammalian cell monolayers to a benznidazole regimen that reduces the intracellular amastigote population to <1% of the pre-treatment level. Of host cells that remained infected, as with the situation in vivo, the vast majority contained only one or two surviving intracellular amastigotes. Analysis, based on non-incorporation of the thymidine analogue EdU, revealed these surviving parasites to be in a transient non-replicative state. Furthermore, treatment with benznidazole led to widespread parasite DNA damage. When the small number of parasites which survive in mice after non-curative treatment were assessed using EdU labelling, this revealed that these persisters were also initially non-replicative. A possible explanation could be that triggering of the T. cruzi DNA damage response pathway by the activity of benznidazole metabolites results in exit from the cell cycle as parasites attempt DNA repair, and that metabolic changes associated with non-proliferation act to reduce drug susceptibility. Alternatively, a small percentage of the parasite population may pre-exist in this non-replicative state prior to treatment.


Asunto(s)
Enfermedad de Chagas , Nitroimidazoles , Parásitos , Tripanocidas , Trypanosoma cruzi , Animales , Ratones , Trypanosoma cruzi/genética , Nitroimidazoles/farmacología , Enfermedad de Chagas/parasitología , Daño del ADN , Tripanocidas/farmacología , Tripanocidas/metabolismo , Mamíferos
9.
Nat Immunol ; 14(5): 514-22, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23563688

RESUMEN

Here we identified B cells as a major source of rapid, innate-like production of interleukin 17 (IL-17) in vivo in response to infection with Trypanosoma cruzi. IL-17(+) B cells had a plasmablast phenotype, outnumbered cells of the TH17 subset of helper T cells and were required for an optimal response to this pathogen. With both mouse and human primary B cells, we found that exposure to parasite-derived trans-sialidase in vitro was sufficient to trigger modification of the cell-surface mucin CD45, which led to signaling dependent on the kinase Btk and production of IL-17A or IL-17F via a transcriptional program independent of the transcription factors RORγt and Ahr. Our combined data suggest that the generation of IL-17(+) B cells may be a previously unappreciated feature of innate immune responses required for pathogen control or IL-17-mediated autoimmunity.


Asunto(s)
Linfocitos B/inmunología , Enfermedad de Chagas/inmunología , Glicoproteínas/metabolismo , Interleucina-17/inmunología , Neuraminidasa/metabolismo , Trypanosoma cruzi/enzimología , Trypanosoma cruzi/inmunología , Animales , Linfocitos B/parasitología , Proliferación Celular , Células Cultivadas , Enfermedad de Chagas/genética , Glicoproteínas/genética , Humanos , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neuraminidasa/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/parasitología , Células Th17/inmunología , Células Th17/parasitología , Activación Transcripcional/inmunología
10.
FASEB J ; 38(6): e23566, 2024 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-38526868

RESUMEN

Trypanosoma cruzi is the causative agent of Chagas disease, a chronic pathology that affects the heart and/or digestive system. This parasite invades and multiplies in virtually all nucleated cells, using a variety of host cell receptors for infection. T. cruzi has a gene that encodes an ecotin-like inhibitor of serine peptidases, ISP2. We generated ISP2-null mutants (Δisp2) in T. cruzi Dm28c using CRISPR/Cas9. Epimastigotes of Δisp2 grew normally in vitro but were more susceptible to lysis by human serum compared to parental and ISP2 add-back lines. Tissue culture trypomastigotes of Δisp2 were more infective to human muscle cells in vitro, which was reverted by the serine peptidase inhibitors aprotinin and camostat, suggesting that host cell epitheliasin/TMPRSS2 is the target of ISP2. Pretreatment of host cells with an antagonist to the protease-activated receptor 2 (PAR2) or an inhibitor of Toll-like receptor 4 (TLR4) selectively counteracted the increased cell invasion by Δisp2, but did not affect invasion by parental and add-back lines. The same was observed following targeted gene silencing of PAR2, TLR4 or TMPRSS2 in host cells by siRNA. Furthermore, Δisp2 caused increased tissue edema in a BALB/c mouse footpad infection model after 3 h differently to that observed following infection with parental and add-back lines. We propose that ISP2 contributes to protect T. cruzi from the anti-microbial effects of human serum and to prevent triggering of PAR2 and TLR4 in host cells, resulting in the modulation of host cell invasion and contributing to decrease inflammation during acute infection.


Asunto(s)
Enfermedad de Chagas , Trypanosoma cruzi , Animales , Ratones , Humanos , Receptor Toll-Like 4/genética , Receptor PAR-2/genética , Enfermedad de Chagas/genética , Enfermedad de Chagas/parasitología , Antivirales/farmacología , Inhibidores de Serina Proteinasa/farmacología , Inflamación , Serina , Serina Endopeptidasas/genética
11.
J Immunol ; 211(7): 1123-1133, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37603014

RESUMEN

Host cell invasion by Trypanosoma cruzi is a markedly silent process, with limited host transcriptional changes indicative of innate immune recognition, except for a modest type I IFN (IFN-I) response. In this study, we show that T. cruzi-induced IFN-ß production was nearly abolished in primary murine cGAS-/- or stimulator of IFN genes (STING)-deficient (STINGGt) macrophages and fibroblasts. T. cruzi infection did not impact the ability of IFN-regulatory factor reporter macrophages to respond to classical cGAS-STING agonists, indicating that the limited IFN-ß induction is not due to active parasite suppression. cGAS-/-, STINGGt, and IFN-α/ß receptor-/- (IFNAR-/-) macrophages infected with T. cruzi yielded significantly higher numbers of amastigotes compared with wild-type macrophages; however, the impact of the STING pathway during infection in vivo is more complex. Despite an initial increase in parasite growth, STINGGt and IFNAR-/- mice ultimately had lower parasite burden in footpads as compared with wild-type mice, demonstrating a role for IFN-I expression in potentiating parasite growth at the infection site. STING pathway activation had little impact on parasite levels in the skeletal muscle; however, in the heart, cGAS-/- and STINGGt mice, but not IFNAR-/- mice, accumulated higher acute parasite loads, suggesting a protective role of STING sensing of T. cruzi in this organ that was independent of IFN-I. Together, these results demonstrate that host cGAS-STING senses T. cruzi infection, enhancing parasite growth at the site of entry, and contributes to acute-phase parasite restriction in the heart, a major site of tissue damage in chronic T. cruzi infection.


Asunto(s)
Enfermedad de Chagas , Interferón Tipo I , Trypanosoma cruzi , Animales , Ratones , Control de Enfermedades Transmisibles , Cromogranina A , Nucleotidiltransferasas/genética
12.
J Immunol ; 210(5): 568-579, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36602929

RESUMEN

T cells are central to the adaptive immune response against Trypanosoma cruzi infection. In chronic Chagas disease (CCD), circulating parasite-specific memory T cells show reduced functionality and increased expression of inhibitory receptors as a result of persistent antigenic stimulation. This phenotype has been linked to progression of cardiac pathology, whereas the presence of polyfunctional T cells shows association with therapeutic success. In this study, we demonstrate that T. cruzi-specific human CD4+ T cells can be identified by their expression of OX40 and CD25 upon in vitro stimulation. We characterized the expression of the inhibitory receptors T cell immunoreceptor with Ig and ITIM domains (TIGIT), T cell Ig and mucin-domain containing-3 (TIM-3), and lymphocyte activation gene 3 (LAG-3) in CD4+ T cells from CCD patients with and without cardiac alterations. Our results show that, independently of their clinical stage, CCD patients present an increased frequency of CD4+ T cells expressing TIGIT in comparison with non-T. cruzi-infected donors. Exposure to parasite Ags increases the expression of TIM-3 in CD4+ T cells from CCD patients, especially in those with cardiac compromise. Upregulation of LAG-3 was also detected in CCD individuals without cardiac manifestations, predominantly within the subpopulation of cells that did not become activated upon stimulation. Further differences were found between groups in the coexpression of these receptors. Blockade of each individual receptor did not affect activation or the production of IFN-γ and IL-10 by CD4+ T cells in response to parasite Ags. Our results suggest a role for TIGIT, TIM-3, and LAG-3 in the modulation of inflammatory phenomena thought to ultimately lead to tissue damage and cardiac pathology.


Asunto(s)
Linfocitos T CD4-Positivos , Enfermedad de Chagas , Humanos , Receptor 2 Celular del Virus de la Hepatitis A , Receptor de Muerte Celular Programada 1/metabolismo , Receptores Inmunológicos
13.
J Immunol ; 210(4): 420-430, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36603035

RESUMEN

Infection with the protozoan parasite Trypanosoma cruzi elicits substantial CD8+ T cell responses that disproportionately target epitopes encoded in the large trans-sialidase (TS) gene family. Within the C57BL/6 infection model, a significant proportion (30-40%) of the T. cruzi-specific CD8+ T cell response targets two immunodominant TS epitopes, TSKb18 and TSKb20. However, both TS-specific CD8+ T cell responses are dispensable for immune control, and TS-based vaccines have no demonstrable impact on parasite persistence, a determinant of disease. Besides TS, the specificity and protective capacity of CD8+ T cells that mediate immune control of T. cruzi infection are unknown. With the goal of identifying alternative CD8+ T cell targets, we designed and screened a representative set of genome-wide, in silico-predicted epitopes. Our screen identified a previously uncharacterized, to our knowledge, T cell epitope MUCKb25, found within mucin family proteins, the third most expanded large gene family in T. cruzi. The MUCKb25-specific response was characterized by delayed kinetics, relative to TS-specific responses, and extensive cross-reactivity with a large number of endogenous epitope variants. Similar to TS-specific responses, the MUCKb25 response was dispensable for control of the infection, and vaccination to generate MUCK-specific CD8+ T cells failed to confer protection. The lack of protection by MUCK vaccination was partly attributed to the fact that MUCKb25-specific T cells exhibit limited recognition of T. cruzi-infected host cells. Overall, these results indicate that the CD8+ T cell compartment in many T. cruzi-infected mice is occupied by cells with minimal apparent effector potential.


Asunto(s)
Enfermedad de Chagas , Vacunas Antiprotozoos , Trypanosoma cruzi , Ratones , Animales , Glicosilfosfatidilinositoles , Mucinas , Señales de Clasificación de Proteína , Ratones Endogámicos C57BL , Linfocitos T CD8-positivos , Epítopos de Linfocito T , Epítopos Inmunodominantes
14.
PLoS Genet ; 18(2): e1010019, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35120121

RESUMEN

Accurate prediction of vectors dispersal, as well as identification of adaptations that allow blood-feeding vectors to thrive in built environments, are a basis for effective disease control. Here we adopted a landscape genomics approach to assay gene flow, possible local adaptation, and drivers of population structure in Rhodnius ecuadoriensis, an important vector of Chagas disease. We used a reduced-representation sequencing technique (2b-RADseq) to obtain 2,552 SNP markers across 272 R. ecuadoriensis samples from 25 collection sites in southern Ecuador. Evidence of high and directional gene flow between seven wild and domestic population pairs across our study site indicates insecticide-based control will be hindered by repeated re-infestation of houses from the forest. Preliminary genome scans across multiple population pairs revealed shared outlier loci potentially consistent with local adaptation to the domestic setting, which we mapped to genes involved with embryogenesis and saliva production. Landscape genomic models showed elevation is a key barrier to R. ecuadoriensis dispersal. Together our results shed early light on the genomic adaptation in triatomine vectors and facilitate vector control by predicting that spatially-targeted, proactive interventions would be more efficacious than current, reactive approaches.


Asunto(s)
Enfermedad de Chagas/epidemiología , Enfermedad de Chagas/genética , Rhodnius/genética , Adaptación Biológica/genética , Animales , Vectores de Enfermedades , Ecosistema , Ecuador/epidemiología , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Flujo Génico , Insectos Vectores/genética , Metagenómica/métodos , Polimorfismo de Nucleótido Simple/genética , Densidad de Población , Rhodnius/patogenicidad , Transcriptoma/genética , Trypanosoma cruzi/genética
15.
J Infect Dis ; 229(1): 198-202, 2024 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-37853514

RESUMEN

BACKGROUND: Chagas disease (CD) is a parasitic disease that affects ∼300 000 people living in the United States. CD leads to cardiac and/or gastrointestinal disease in up to 30% of untreated people. However, end-organ damage can be prevented with early diagnosis and antiparasitic therapy. METHODS: We reviewed electronic health records of patients who underwent testing for CD at four hospital systems in California and Texas between 2016 and 2020. Descriptive analyses were performed as a needs assessment for improving CD diagnosis. RESULTS: In total, 470 patients were tested for CD. Cardiac indications made up more than half (60%) of all testing, and the most frequently cited cardiac condition was heart failure. Fewer than 1% of tests were ordered by obstetric and gynecologic services. Fewer than half (47%) of patients had confirmatory testing performed at the Centers for Disease Control and Prevention. DISCUSSION: Four major hospitals systems in California and Texas demonstrated low overall rates of CD diagnostic testing, testing primarily among older patients with end-organ damage, and incomplete confirmatory testing. This suggests missed opportunities to diagnose CD in at-risk individuals early in the course of infection when antiparasitic treatment can reduce the risk of disease progression and prevent vertical transmission.


Asunto(s)
Enfermedad de Chagas , Trypanosoma cruzi , Embarazo , Humanos , Femenino , Estados Unidos , Texas/epidemiología , Enfermedad de Chagas/diagnóstico , Enfermedad de Chagas/tratamiento farmacológico , Enfermedad de Chagas/epidemiología , California/epidemiología , Antiparasitarios
16.
J Biol Chem ; 299(6): 104715, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37061002

RESUMEN

Trypanosomatids are a diverse group of uniflagellate protozoan parasites that include globally relevant pathogens such as Trypanosoma cruzi, the causative agent of Chagas disease. Trypanosomes lack the fatty acid synthase system typically used for de novo fatty acid (FA) synthesis in other eukaryotes. Instead, these microbes have evolved a modular FA elongase (ELO) system comprised of individual ELO enzymes (ELO1-4) that can operate processively to generate long chain- and very long chain-FAs. The importance of ELO's for maintaining lipid homeostasis in trypanosomatids is currently unclear, given their ability to take up and utilize exogenous FAs for lipid synthesis. To assess ELO function in T. cruzi, we generated individual KO lines, Δelo1, Δelo2, and Δelo3, in which the genes encoding ELO1-3 were functionally disrupted in the parasite insect stage (epimastigote). Using unbiased lipidomic and metabolomic analyses, in combination with metabolic tracing and biochemical approaches, we demonstrate that ELO2 and ELO3 are required for global lipid homeostasis, whereas ELO1 is dispensable for this function. Instead, ELO1 activity is needed to sustain mitochondrial activity and normal growth in T. cruzi epimastigotes. The cross-talk between microsomal ELO1 and the mitochondrion is a novel finding that, we propose, merits further examination of the trypanosomatid ELO pathway as critical for central metabolism.


Asunto(s)
Enfermedad de Chagas , Trypanosoma cruzi , Humanos , Trypanosoma cruzi/genética , Trypanosoma cruzi/metabolismo , Elongasas de Ácidos Grasos/metabolismo , Enfermedad de Chagas/genética , Enfermedad de Chagas/metabolismo , Homeostasis , Mitocondrias/genética , Mitocondrias/metabolismo , Lípidos
17.
J Biol Chem ; 299(7): 104857, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37230387

RESUMEN

The TcK2 protein kinase of Trypanosoma cruzi, the causative agent of Chagas disease, is structurally similar to the human kinase PERK, which phosphorylates the initiation factor eIF2α and, in turn, inhibits translation initiation. We have previously shown that absence of TcK2 kinase impairs parasite proliferation within mammalian cells, positioning it as a potential target for treatment of Chagas disease. To better understand its role in the parasite, here we initially confirmed the importance of TcK2 in parasite proliferation by generating CRISPR/Cas9 TcK2-null cells, albeit they more efficiently differentiate into infective forms. Proteomics indicates that the TcK2 knockout of proliferative forms expresses proteins including trans-sialidases, normally restricted to infective and nonproliferative trypomastigotes explaining decreased proliferation and better differentiation. TcK2 knockout cells lost phosphorylation of eukaryotic initiation factor 3 and cyclic AMP responsive-like element, recognized to promote growth, likely explaining both decreased proliferation and augmented differentiation. To identify specific inhibitors, a library of 379 kinase inhibitors was screened by differential scanning fluorimetry using a recombinant TcK2 encompassing the kinase domain and selected molecules were tested for kinase inhibition. Only Dasatinib and PF-477736, inhibitors of Src/Abl and ChK1 kinases, showed inhibitory activity with IC50 of 0.2 ± 0.02 mM and 0.8 ± 0.1, respectively. In infected cells Dasatinib inhibited growth of parental amastigotes (IC50 = 0.6 ± 0.2 mM) but not TcK2 of depleted parasites (IC50 > 34 mM) identifying Dasatinib as a potential lead for development of therapeutics for Chagas disease targeting TcK2.


Asunto(s)
Enfermedad de Chagas , Parásitos , Trypanosoma cruzi , Animales , Humanos , Trypanosoma cruzi/genética , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo , Dasatinib , Enfermedad de Chagas/tratamiento farmacológico , Enfermedad de Chagas/parasitología , Proliferación Celular , Mamíferos/metabolismo
18.
Clin Infect Dis ; 78(2): 453-456, 2024 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-37805935

RESUMEN

Chagas disease (CD), caused by Trypanosoma cruzi, is underdiagnosed in the United States. Improved screening strategies are needed, particularly for people at risk for life-threatening sequelae of CD, including people with human immunodeficiency virus (HIV, PWH). Here we report results of a CD screening strategy applied at a large HIV clinic serving an at-risk population.


Asunto(s)
Enfermedad de Chagas , Infecciones por VIH , Trypanosoma cruzi , Humanos , Estados Unidos/epidemiología , VIH , Enfermedad de Chagas/diagnóstico , Enfermedad de Chagas/epidemiología , Enfermedad de Chagas/complicaciones , Infecciones por VIH/diagnóstico , Infecciones por VIH/epidemiología , Infecciones por VIH/complicaciones
19.
Clin Infect Dis ; 78(Supplement_2): S175-S182, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38662705

RESUMEN

BACKGROUND: Neglected tropical diseases are responsible for considerable morbidity and mortality in low-income populations. International efforts have reduced their global burden, but transmission is persistent and case-finding-based interventions rarely target asymptomatic individuals. METHODS: We develop a generic mathematical modeling framework for analyzing the dynamics of visceral leishmaniasis in the Indian sub-continent (VL), gambiense sleeping sickness (gHAT), and Chagas disease and use it to assess the possible contribution of asymptomatics who later develop disease (pre-symptomatics) and those who do not (non-symptomatics) to the maintenance of infection. Plausible interventions, including active screening, vector control, and reduced time to detection, are simulated for the three diseases. RESULTS: We found that the high asymptomatic contribution to transmission for Chagas and gHAT and the apparently high basic reproductive number of VL may undermine long-term control. However, the ability to treat some asymptomatics for Chagas and gHAT should make them more controllable, albeit over relatively long time periods due to the slow dynamics of these diseases. For VL, the toxicity of available therapeutics means the asymptomatic population cannot currently be treated, but combining treatment of symptomatics and vector control could yield a quick reduction in transmission. CONCLUSIONS: Despite the uncertainty in natural history, it appears there is already a relatively good toolbox of interventions to eliminate gHAT, and it is likely that Chagas will need improvements to diagnostics and their use to better target pre-symptomatics. The situation for VL is less clear, and model predictions could be improved by additional empirical data. However, interventions may have to improve to successfully eliminate this disease.


Asunto(s)
Infecciones Asintomáticas , Enfermedad de Chagas , Leishmaniasis Visceral , Modelos Teóricos , Enfermedades Desatendidas , Humanos , Enfermedades Desatendidas/prevención & control , Enfermedades Desatendidas/epidemiología , Enfermedad de Chagas/transmisión , Enfermedad de Chagas/prevención & control , Enfermedad de Chagas/epidemiología , Enfermedad de Chagas/tratamiento farmacológico , Infecciones Asintomáticas/epidemiología , Leishmaniasis Visceral/prevención & control , Leishmaniasis Visceral/epidemiología , Leishmaniasis Visceral/transmisión , Leishmaniasis Visceral/tratamiento farmacológico , Tripanosomiasis Africana/prevención & control , Tripanosomiasis Africana/epidemiología , Tripanosomiasis Africana/transmisión , Tripanosomiasis Africana/tratamiento farmacológico , India/epidemiología , Animales
20.
Am J Pathol ; 193(3): 313-331, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36565805

RESUMEN

Chagas disease is caused by Trypanosoma cruzi. This study aimed to determine the effects of T. cruzi infection on fertility rate and health of the newborn pups in pregnant mice. Female mice were challenged with T. cruzi and mated at 21 days (acute parasitemic phase) or 90 days (chronic parasite persistence phase) after infection. Pups were examined for growth up to 20 days after birth; and parasite burden in brain, heart, skeletal muscle, and intestine was measured by real-time quantitative PCR. The inflammatory infiltrate, necrosis, and fibrosis in pups' heart and brain tissues were evaluated by histology. T. cruzi infection in dams delayed the onset of pregnancy, decreased the fertility rate, and led to vertical transmission of parasite to the pups. Furthermore, infected dams delivered pups that exhibited decreased survival rate, decreased birth weight, and decreased growth rate. Significantly increased inflammation, necrosis, and fibrosis of cardiac and brain tissues were noted in pups born to infected dams. Initial challenge with higher parasite dose had more detrimental effects on fertility rate and pups' health in both acutely and chronically infected dams. In conclusion, mice offer a promising model to evaluate the efficacy of new vaccines and therapeutic drugs in controlling the acute and chronic maternal T. cruzi infection and congenital transmission to newborns, and in improving the fertility rate and pups' health outcomes.


Asunto(s)
Enfermedad de Chagas , Parásitos , Trypanosoma cruzi , Embarazo , Femenino , Ratones , Animales , Resultado del Embarazo , Enfermedad de Chagas/parasitología , Fibrosis , Necrosis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA