Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 201
Filter
1.
Parasite Immunol ; 46(5): e13036, 2024 May.
Article in English | MEDLINE | ID: mdl-38720445

ABSTRACT

Apolipoprotein E (ApoE) has been associated with several diseases including Parkinson's disease, Alzheimer's and multiple sclerosis. ApoE also has documented immunomodulatory functions. We investigated gene expression in circulating monocytes and in bone marrows of patients with visceral leishmaniasis (VL) living in an endemic area in Bihar, India, and contrasted these with control healthy subjects or other diagnostic bone marrows from individuals in the same region. Samples from VL patients were obtained prior to initiating treatment. Our study revealed significant upregulated expression of the apoE transcript in patients with VL. Furthermore, the levels of ApoE protein were elevated in serum samples of subjects with VL compared with healthy endemic controls. These observations may provide clues regarding the complex interactions between lipid metabolism and immunoregulation of infectious and inflammatory diseases.


Subject(s)
Apolipoproteins E , Leishmaniasis, Visceral , Monocytes , Up-Regulation , Adolescent , Adult , Child , Female , Humans , Male , Middle Aged , Young Adult , Apolipoproteins E/genetics , Bone Marrow , India/epidemiology , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/blood , Leishmaniasis, Visceral/parasitology , Monocytes/immunology
2.
PLoS Negl Trop Dis ; 18(4): e0012112, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38669292

ABSTRACT

Visceral leishmaniasis (VL) is a potentially fatal parasitic infection caused by Leishmania donovani in India. L. donovani is an obligate intracellular protozoan residing mostly in macrophages of the reticuloendothelial system throughout chronic infection. Monocytic phagocytes are critical in the pathogenesis of different forms of leishmaniasis. Subsets of monocytes are distinguished by their surface markers into CD14+CD16- classical monocytes, CD14+CD16+ intermediate monocytes, and CD16++CD14low non-classical monocyte subsets. During cutaneous leishmaniasis (CL), intermediate monocyte are reported to be a source of inflammatory cytokines IL-1ß and TNF, and they express CCR2 attracting them to sites of inflammatory pathology. We examined monocyte subsets in the blood and bone marrow of patients with VL from an endemic site in Bihar, India, and found these contrasted with the roles of monocytes in CL. During VL, intermediate and non-classical CD16+ monocyte subsets expressed instead a non-inflammatory phenotype with low CCR2, high CX3CR1 and low microbicidal oxidant generation, making them more similar to patrolling monocytes than inflammatory cells. Bone marrow CD16+ monocyte subsets expressed a phenotype that might be more similar to the inflammatory subsets of CL, although our inability to obtain bone marrow from healthy donors in the endemic region hampered this interpretation Overall the data suggest that CD16+ intermediate monocyte subsets in VL patients express a phenotypes that contributes to an immunosuppressed pathologic immune state, but in contrast to CL, these do not mediate localized inflammatory responses.


Subject(s)
Bone Marrow , Leishmaniasis, Visceral , Monocytes , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/parasitology , Humans , Monocytes/immunology , India , Adult , Male , Bone Marrow/parasitology , Female , Receptors, IgG/analysis , Receptors, IgG/metabolism , Leishmania donovani/immunology , Leishmania donovani/physiology , Young Adult , Adolescent , Receptors, CCR2/metabolism , Middle Aged , Child , Receptors, Chemokine/metabolism , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Cytokines/metabolism
3.
PLoS Negl Trop Dis ; 17(4): e0011206, 2023 04.
Article in English | MEDLINE | ID: mdl-37011128

ABSTRACT

Visceral leishmaniasis (VL) is a neglected tropical disease that is globally distributed and has the potential to cause very serious illness. Prior literature highlights the emergence and spread of VL is influenced by multiple factors, such as socioeconomic status, sanitation levels or animal and human reservoirs. The study aimed to retrospectively investigate the presence and infectiousness of VL in Rio Grande do Norte (RN), Brazil between 2007 and 2020. We applied a hierarchical Bayesian approach to estimate municipality-specific relative risk of VL across space and time. The results show evidence that lower socioeconomic status is connected to higher municipality-specific VL risk. Overall, estimates reveal spatially heterogeneous VL risks in RN, with a high probability that VL risk for municipalities within the West Potiguar mesoregion are more than double the expected VL risk. Additionally, given the data available, results indicate there is a high probability of increasing VL risk in the municipalities of Natal, Patu and Pau dos Ferros. These findings demonstrate opportunities for municipality-specific public health policy interventions and warrant future research on identifying epidemiological drivers in at-risk regions.


Subject(s)
Leishmaniasis, Visceral , Animals , Humans , Leishmaniasis, Visceral/epidemiology , Retrospective Studies , Brazil/epidemiology , Bayes Theorem , Cities , Neglected Diseases
4.
medRxiv ; 2023 Mar 06.
Article in English | MEDLINE | ID: mdl-36945413

ABSTRACT

Background: Leishmania infantum is an opportunistic parasitic infection. An immunocompromised state increases the risk of converting asymptomatic infection to symptomatic visceral leishmaniasis (VL), which has a ~5% fatality rate even with treatment. HIV coinfection increases the risk of death from VL. Methods: A cross-sectional study was performed between 2014 and 2016 to determine the prevalence of L. infantum infection in HIV positive subjects residing in the state of Rio Grande do Norte, Brazil (n=1,372) and of these a subgroup of subjects were followed longitudinally. Subsequent incident cases of VL were ascertained from a public health database through 2018. A subgroup (n=69) of the cross-sectional study subjects was chosen to assess immune status (T cell activation, senescence, exhaustion) and outcome. The data were compared between asymptomatic HIV+/L. infantum+ (HIV/Leish), symptomatic visceral leishmaniasis (VL), recovered VL, DTH+ (Delayed-Type Hypersensitivity response - Leishmanin skin test), AIDS/VL, HIV+ only (HIV+), and Non-HIV/Non L. infantum infection (control subjects). Results: The cross-sectional study showed 24.2% of HIV+ subjects had positive anti-IgG Leishmania antibodies. After 3 years, 2.4% (8 of 333) of these HIV/Leish coinfected subjects developed AIDS/VL, whereas 1.05% (11 of 1,039) of HIV subjects with negative leishmania serology developed AIDS/VL. Poor adherence to antiretroviral therapy (p=0.0008) or prior opportunistic infections (p=0.0007) was associated with development of AIDS/VL. CD4+ (p=0.29) and CD8+ (p=0.38) T cells counts or viral load (p=0.34) were similar between asymptomatic HIV/Leish and HIV subjects. However, activated CD8+CD38+HLA-DR+ T cells were higher in asymptomatic HIV/Leish than HIV group. Likewise, senescent (CD57+) or exhausted (PD1+) CD8+ T cells were higher in asymptomatic HIV/Leish than in AIDS/VL or HIV groups. Conclusion: Although asymptomatic HIV/Leish subjects had normal and similar CD4+ and CD8+ T cells counts, their CD8+T cells had increased activation, senescence, and exhaustion, which could contribute to risk of developing VL.

5.
PLoS Negl Trop Dis ; 16(6): e0010390, 2022 06.
Article in English | MEDLINE | ID: mdl-35704664

ABSTRACT

BACKGROUND: Leishmaniases are neglected tropical diseases that inflict great burden to poor areas of the globe. Intense research has aimed to identify parasite genetic signatures predictive of infection outcomes. Consistency of diagnostic tools based on these markers would greatly benefit from accurate understanding of Leishmania spp. population genetics. We explored two chromosomal loci to characterize a population of L. braziliensis causing human disease in Northeast Brazil. METHODOLOGY/PRINCIPAL FINDINGS: Two temporally distinct samples of L. braziliensis were obtained from patients attending the leishmaniasis clinic at the village of Corte de Pedra: (2008-2011) primary sample, N = 120; (1999-2001) validation sample, N = 35. Parasites were genotyped by Sanger's sequencing of two 600 base pairs loci starting at nucleotide positions 3,074 and 425,451 of chromosomes 24 and 28, respectively. Genotypes based on haplotypes of biallelic positions in each locus were tested for several population genetic parameters as well as for geographic clustering within the region. Ample geographic overlap of genotypes at the two loci was observed as indicated by non-significant Cusick and Edward's comparisons. No linkage disequilibrium was detected among combinations of haplotypes for both parasite samples. Homozygous and heterozygous genotypes displayed Hardy-Weinberg equilibrium (HWE) at both loci in the two samples when straight observed and expected counts were compared by Chi-square (p>0.5). However, Bayesian statistics using one million Monte-Carlo randomizations disclosed a less robust HWE for chromosome 24 genotypes, particularly in the primary sample (p = 0.04). Fixation indices (Fst) were consistently lower than 0.05 among individuals of the two samples at both tested loci, and no intra-populational structuralization could be detected using STRUCTURE software. CONCLUSIONS/SIGNIFICANCE: These findings suggest that L. braziliensis can maintain stable populations in foci of human leishmaniasis and are capable of robust genetic recombination possibly due to events of sexual reproduction during the parasite's lifecycle.


Subject(s)
Leishmania braziliensis , Leishmaniasis, Cutaneous , Leishmaniasis , Bayes Theorem , Brazil/epidemiology , Genotype , Humans , Leishmania braziliensis/genetics , Leishmaniasis/parasitology , Leishmaniasis, Cutaneous/epidemiology , Leishmaniasis, Cutaneous/parasitology
7.
Curr Opin Infect Dis ; 34(5): 409-414, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34267043

ABSTRACT

PURPOSE OF REVIEW: The current article will review how the coronavirus disease 2019 pandemic has changed travel and travel medicine. RECENT FINDINGS: Travelers spread severe acute respiratory syndrome coronavirus 2 globally and continue to spread variants. The characteristics of the virus, the place, and time created a perfect storm that allowed the virus to quickly spread globally. The virus spread by every mode of travel with risk of transmission influenced by proximity to an infected person, duration of trip, physical characteristics of the space, and ventilation. Superspreading events were common; a small percentage of infected people accounted for most of transmission. The travel and tourist industry was devastated as lockdowns and quarantines severely restricted domestic and international travel. A trip includes multiple segments and shared sequential spaces, mostly indoors. Creating safe travel requires attention to all segments of a trip. SUMMARY: The coronavirus disease 2019 pandemic has affected every part of travel and travel medicine. The rapid development of multiple safe and effective vaccines and their deployment is allowing resumption of travel, yet many populations lack access to vaccines, and high levels of transmission continue in many areas. Providing documentation of vaccination or immunity in a consistent, verifiable, interoperable system is one of many active issues.


Subject(s)
COVID-19/immunology , Pandemics/prevention & control , Animals , Communicable Disease Control/methods , Humans , SARS-CoV-2/immunology , Travel , Travel Medicine/methods , Vaccination/methods
8.
mSphere ; 6(4): e0042321, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34259561

ABSTRACT

Visceral leishmaniasis is a potentially fatal disease caused by the protozoon Leishmania donovani or L. infantum (Li). Although previous studies revealed that high lipid intake reduces parasite burdens in Leishmania donovani-infected mice, the specific contributions of dietary lipids to Li-associated pathogenesis are not known. To address this, we evaluated parasite growth, liver pathology, and transcriptomic signatures in Li-infected BALB/c mice fed either a control, high-fat, high-cholesterol, or high-fat-high-cholesterol diet. Using quantitative PCR (qPCR), we observed significantly reduced liver parasite burdens in mice fed the high-fat-high-cholesterol diet compared to mice fed the control diet. In contrast to the liver, parasite expansion occurred earlier in the spleens of mice fed the experimental diets. Histological examination revealed an intense inflammatory cell infiltrate in livers predominantly composed of neutrophils caused by the high-fat-high-cholesterol diet specifically. After 8 weeks of infection (12 weeks of diet), Illumina microarrays revealed significantly increased expression of transcripts belonging to immune- and angiogenesis-related pathways in livers of both uninfected and Li-infected mice fed the high-fat-high-cholesterol diet. These data suggest that increased fat and cholesterol intake prior to Li infection leads to a hepatic inflammatory environment and thus reduces the parasite burden in the liver. Defining inflammatory signatures as well as pathology in the liver may reveal opportunities to modify the therapeutic approach to Li infection. IMPORTANCE Leishmaniasis is a spectrum of diseases caused by Leishmania species protozoa that is most common in warm climates, coinciding with impoverished regions. Visceral leishmaniasis is a potentially fatal disease in which parasites infect reticuloendothelial organs and cause progressive wasting and immunocompromise. The distribution and demographics of visceral leishmaniasis have changed over recent years, coinciding with modernizing societies and the increased availability of Western diets rich in lipid content. We report here that increased dietary fat and cholesterol intake affected disease pathogenesis by increasing inflammation and reducing localized parasite burdens in the liver. These diet-induced changes in disease pathogenesis might explain in part the changing epidemiology of visceral leishmaniasis. A relationship between diet and inflammatory responses may occur in leishmaniasis and other microbial or immune-mediated diseases, possibly revealing opportunities to modify the therapeutic approach to microbial infections.


Subject(s)
Dietary Fats/metabolism , Inflammation/complications , Leishmania infantum/growth & development , Leishmaniasis, Visceral/parasitology , Animal Feed/analysis , Animals , Female , Inflammation/immunology , Leishmania infantum/genetics , Leishmania infantum/metabolism , Leishmaniasis, Visceral/immunology , Liver/immunology , Liver/parasitology , Metabolic Networks and Pathways , Mice , Mice, Inbred BALB C , Parasite Load , Spleen/immunology , Spleen/parasitology
9.
J Exp Med ; 218(6)2021 06 07.
Article in English | MEDLINE | ID: mdl-33830176

ABSTRACT

Antimalarial antibody responses are essential for mediating the clearance of Plasmodium parasite-infected RBCs from infected hosts. However, the rapid appearance of large numbers of plasmablasts in Plasmodium-infected hosts can suppress the development and function of durable humoral immunity. Here, we identify that the formation of plasmablast populations in Plasmodium-infected mice is mechanistically linked to both hemolysis-induced exposure of phosphatidylserine on damaged RBCs and inflammatory cues. We also show that virus and Trypanosoma infections known to trigger hemolytic anemia and high-grade inflammation also induce exuberant plasmablast responses. The induction of hemolysis or administration of RBC membrane ghosts increases plasmablast differentiation. The phosphatidylserine receptor Axl is critical for optimal plasmablast formation, and blocking phosphatidylserine limits plasmablast expansions and reduces Plasmodium parasite burden in vivo. Our findings support that strategies aimed at modulating polyclonal B cell activation and phosphatidylserine exposure may improve immune responses against Plasmodium parasites and potentially other infectious diseases that are associated with anemia.


Subject(s)
Cell Differentiation/immunology , Hemolysis/immunology , Phosphatidylserines/immunology , Plasma Cells/immunology , Animals , Antibodies, Protozoan/immunology , Antimalarials/immunology , B-Lymphocytes/immunology , B-Lymphocytes/parasitology , Cells, Cultured , Erythrocytes/immunology , Erythrocytes/parasitology , Humans , Immunity, Humoral/immunology , Malaria/immunology , Malaria/parasitology , Mice , Mice, Inbred C57BL , Plasma Cells/parasitology , Plasmodium yoelii/immunology
10.
Exp Parasitol ; 220: 107968, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32781093

ABSTRACT

The parasitic protozoan Leishmania infantum resides primarily in macrophages throughout mammalian infection. Infection is initiated by deposition of the metacyclic promastigote into the dermis of a mammalian host by the sand fly vector. Promastigotes enter macrophages by ligating surface receptors such as complement receptor 3 (CR3), inducing phagocytosis of the parasite. At the binding site of metacyclic promastigotes, we observed large asymmetrical aggregates of macrophage membrane with underlying actin, resembling membrane ruffles. Actin accumulation was observed at the point of initial contact, before phagosome formation and accumulation of peri-phagosomal actin. Ruffle-like structures did not form during phagocytosis of attenuated promastigotes or during phagocytosis of the intracellular amastigote form of L. infantum. Entry of promastigotes through massive actin accumulation was associated with a subsequent delay in fusion of the parasitophorous vacuole (PV) with the lysosomal markers LAMP-1 and Cathepsin D. Actin accumulation was also associated with entry through CR3, since macrophages from CD11b knockout (KO) mice did not form massive aggregates of actin during phagocytosis of metacyclic promastigotes. Furthermore, intracellular survival of L. infantum was significantly decreased in CD11b KO compared to wild type macrophages, although entry rates were similar. We conclude that both promastigote virulence and host cell CR3 are needed for the formation of ruffle-like membrane structures at the site of metacyclic promastigote phagocytosis, and that formation of actin-rich aggregates during entry correlates with the intracellular survival of virulent promastigotes.


Subject(s)
Actins/metabolism , Leishmania infantum/physiology , Leishmaniasis, Visceral/parasitology , Macrophage-1 Antigen/physiology , Phagocytosis/physiology , Animals , Cathepsin D/metabolism , Cell Membrane/ultrastructure , Cricetinae , Humans , Leishmania infantum/pathogenicity , Leishmania infantum/ultrastructure , Lysosomal-Associated Membrane Protein 1/metabolism , Macrophages/parasitology , Male , Mesocricetus , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Confocal , Vacuoles/parasitology , Virulence
11.
J Infect Dis ; 223(3): 517-521, 2021 02 13.
Article in English | MEDLINE | ID: mdl-32614452

ABSTRACT

Visceral leishmaniasis (VL; Leishmania donovani) cases produce interferon-γ and tumor necrosis factor in response to soluble leishmanial antigen (SLA) in whole-blood assays. Using transcriptional profiling, we demonstrate the impact of interleukin-10 (IL-10), a cytokine implicated in VL, on this response. SLA stimulation identified 28 differentially expressed genes (DEGs), 17/28 in a single network with TNF as hub. SLA plus anti-IL-10 produced 454 DEGs, 292 in a single network with TNF, IFNG, NFKBIA, IL6, and IL1B as hubs in concert with a remarkable chemokine/cytokine storm. Our data demonstrate the singular effect of IL-10 as a potent immune modulator in VL.


Subject(s)
Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Interleukin-10/immunology , Leishmaniasis, Visceral/immunology , Antibodies, Protozoan/blood , Antibodies, Protozoan/genetics , Antigens, Protozoan/blood , Cytokines/immunology , Gene Expression , Humans , Interferon-gamma/immunology , Leishmania donovani/immunology , Tumor Necrosis Factor-alpha
17.
Article in English | MEDLINE | ID: mdl-31938550

ABSTRACT

Yellow fever (YF) outbreaks continue, have expanded into new areas and threaten large populations in South America and Africa. Predicting where epidemics might occur must take into account local mosquito populations and specific YF virus strain, as well as ecoclimatic conditions, sociopolitical and demographic factors including population size, density, and mobility, and vaccine coverage. Populations of Aedes aegypti and Aedes albopictus from different regions vary in susceptibility to and capacity to transmit YF virus. YF virus cannot be eliminated today because the virus circulates in animal reservoirs, but human disease could be eliminated with wide use of the vaccine. WHO EYE (Eliminate Yellow Fever Epidemics) is a welcome plan to control YF, with strategies to be carried out from 2017 to 2026: to expand use of YF vaccine, to prevent international spread, and to contain outbreaks rapidly. YF vaccination is the mainstay in controlling YF outbreaks, but global supply is insufficient. Therefore, dose-sparing strategies have been proposed including fractional dosing and intradermal administration. Fractional dosing has been effectively used in outbreak control but currently does not satisfy International Health Regulations; special documentation is needed for international travel. Vector control is another facet in preventing YF outbreaks, and novel methods are being considered and proposed.

19.
Biometrics ; 76(3): 711-721, 2020 09.
Article in English | MEDLINE | ID: mdl-31785149

ABSTRACT

Zoonotic visceral leishmaniasis (ZVL) is a serious neglected tropical disease that is endemic in 98 countries. ZVL is primarily transmitted via a sand fly vector. In the United States, it is enzootic in some canine populations; it is transmitted from infectious mother to pup transplacentally, and vector-borne transmission is absent. This absence affords a unique opportunity to study (1) vertical transmission dynamics in dogs and (2) the importance of vertical transmission in maintaining an infectious reservoir in the presence of a vector. In this paper, we present Bayesian compartmental models and reproductive number formulations to examine (1) and (2), providing a mechanism to plan and evaluate interventions in regions where both transmission modes are present. First, we propose an individual-level susceptible, infectious, removed (SIR) model to study the effect of maternal infection status during pregnancy on pup infection progression. We provide evidence that pups born to diagnostically positive mothers during pregnancy are more likely to become diagnostically positive both earlier in life, and at some point during their lifetime, than those born to diagnostically negative mothers. Second, we propose a population-level SIR model to study the impact of a vertically maintained reservoir on propagating infection in a naive canine population through emergent vector transmission using simulation studies. We also present reproductive numbers to quantify contributions of vertically infected and vector-infected dogs to maintaining infection in the population. We show that a vertically maintained canine reservoir can propagate infection in a theoretical naive population in the presence of a vector.


Subject(s)
Dog Diseases , Leishmaniasis, Visceral , Animals , Bayes Theorem , Computer Simulation , Dogs , Female , Infectious Disease Transmission, Vertical , Leishmaniasis, Visceral/veterinary , Pregnancy , United States
20.
J Appl Stat ; 46(6): 1043-1065, 2019.
Article in English | MEDLINE | ID: mdl-31537954

ABSTRACT

Population-level proportions of individuals that fall at different points in the spectrum [of disease severity], from asymptomatic infection to severe disease, are often difficult to observe, but estimating these quantities can provide information about the nature and severity of the disease in a particular population. Logistic and multinomial regression techniques are often applied to infectious disease modeling of large populations and are suited to identifying variables associated with a particular disease or disease state. However, they are less appropriate for estimating infection state prevalence over time because they do not naturally accommodate known disease dynamics like duration of time an individual is infectious, heterogeneity in the risk of acquiring infection, and patterns of seasonality. We propose a Bayesian compartmental model to estimate latent infection state prevalence over time that easily incorporates known disease dynamics. We demonstrate how and why a stochastic compartmental model is a better approach for determining infection state proportions than multinomial regression is by using a novel method for estimating Bayes factors for models with high-dimensional parameter spaces. We provide an example using visceral leishmaniasis in Brazil and present an empirically-adjusted reproductive number for the infection.

SELECTION OF CITATIONS
SEARCH DETAIL
...