Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
EBioMedicine ; 86: 104378, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36462405

ABSTRACT

BACKGROUND: Miltefosine treatment failure in visceral leishmaniasis in Brazil has been associated with deletion of the miltefosine susceptibility locus (MSL) in Leishmania infantum. The MSL comprises four genes, 3'-nucleotidase/nucleases (NUC1 and NUC2); helicase-like protein (HLP); and 3,2-trans-enoyl-CoA isomerase (TEI). METHODS: In this study CRISPR-Cas9 was used to either epitope tag or delete NUC1, NUC2, HLP and TEI, to investigate their role in miltefosine resistance mechanisms. Additionally, miltefosine transporter genes and miltefosine-mediated reactive oxygen species homeostasis were assessed in 26 L. infantum clinical isolates. A comparative lipidomic analysis was also performed to investigate the molecular basis of miltefosine resistance. FINDINGS: Deletion of both NUC1, NUC2 from the MSL was associated with a significant decrease in miltefosine susceptibility, which was restored after re-expression. Metabolomic analysis of parasites lacking the MSL or NUC1 and NUC2 identified an increase in the parasite lipid content, including ergosterol; these lipids may contribute to miltefosine resistance by binding the drug in the membrane. Parasites lacking the MSL are more resistant to lipid metabolism perturbation caused by miltefosine and NUC1 and NUC2 are involved in this pathway. Additionally, L. infantum parasites lacking the MSL isolated from patients who relapsed after miltefosine treatment were found to modulate nitric oxide accumulation in host macrophages. INTERPRETATION: Altogether, these data indicate that multifactorial mechanisms are involved in natural resistance to miltefosine in L. infantum and that the absence of the 3'nucleotidase/nuclease genes NUC1 and NUC2 contributes to the phenotype. FUNDING: MRC GCRF and FAPES.


Subject(s)
Antiprotozoal Agents , Leishmania infantum , Leishmania infantum/genetics , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/therapeutic use , Phosphorylcholine/pharmacology , Phosphorylcholine/therapeutic use , Nucleotidases/metabolism
2.
Front Immunol ; 12: 700501, 2021.
Article in English | MEDLINE | ID: mdl-34557190

ABSTRACT

Ly6Chi inflammatory monocytes develop in the bone marrow and migrate to the site of infection during inflammation. Upon recruitment, Ly6Chi monocytes can differentiate into dendritic cells or macrophages. According to the tissue environment they can also acquire different functions. Several studies have described pre-activation of Ly6Chi monocytes in the bone marrow during parasitic infection, but whether this process occurs during experimental visceral leishmaniasis and, if so, the mechanisms contributing to their activation are yet to be established. In wild type C57BL/6 (B6) mice infected with Leishmania donovani, the number of bone marrow Ly6Chi monocytes increased over time. Ly6Chi monocytes displayed a highly activated phenotype from 28 days to 5 months post infection (p.i), with >90% expressing MHCII and >20% expressing iNOS. In comparison, in B6.Rag2-/- mice <10% of bone marrow monocytes were MHCII+ at day 28 p.i., an activation deficiency that was reversed by adoptive transfer of CD4+ T cells. Depletion of CD4+ T cells in B6 mice and the use of mixed bone marrow chimeras further indicated that monocyte activation was driven by IFNγ produced by CD4+ T cells. In B6.Il10-/- mice, L. donovani infection induced a faster but transient activation of bone marrow monocytes, which correlated with the magnitude of CD4+ T cell production of IFNγ and resolution of the infection. Under all of the above conditions, monocyte activation was associated with greater control of parasite load in the bone marrow. Through reinfection studies in B6.Il10-/- mice and drug (AmBisome®) treatment of B6 mice, we also show the dependence of monocyte activation on parasite load. In summary, these data demonstrate that during L. donovani infection, Ly6Chi monocytes are primed in the bone marrow in a process driven by CD4+ T cells and whereby IFNγ promotes and IL-10 limits monocyte activation and that the presence of parasites/parasite antigen plays a crucial role in maintaining bone marrow monocyte activation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Interferon-gamma/immunology , Leishmaniasis, Visceral/immunology , Monocytes/immunology , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL
3.
Front Immunol ; 12: 795554, 2021.
Article in English | MEDLINE | ID: mdl-34975901

ABSTRACT

Increasing evidence suggests that in hosts infected with parasites of the Leishmania donovani complex, transmission of infection to the sand fly vector is linked to parasite repositories in the host skin. However, a detailed understanding of the dispersal (the mechanism of spread) and dispersion (the observed state of spread) of these obligatory-intracellular parasites and their host phagocytes in the skin is lacking. Using endogenously fluorescent parasites as a proxy, we apply image analysis combined with spatial point pattern models borrowed from ecology to characterize dispersion of parasitized myeloid cells (including ManR+ and CD11c+ cells) and predict dispersal mechanisms in a previously described immunodeficient model of L. donovani infection. Our results suggest that after initial seeding of infection in the skin, heavily parasite-infected myeloid cells are found in patches that resemble innate granulomas. Spread of parasites from these initial patches subsequently occurs through infection of recruited myeloid cells, ultimately leading to self-propagating networks of patch clusters. This combination of imaging and ecological pattern analysis to identify mechanisms driving the skin parasite landscape offers new perspectives on myeloid cell behavior following parasitism by L. donovani and may also be applicable to elucidating the behavior of other intracellular tissue-resident pathogens and their host cells.


Subject(s)
Image Processing, Computer-Assisted , Leishmania donovani/pathogenicity , Leishmaniasis, Visceral/parasitology , Microscopy, Confocal , Microscopy, Fluorescence , Myeloid Cells/parasitology , Skin/parasitology , Spatial Analysis , Animals , CD11 Antigens/metabolism , Cluster Analysis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Host-Parasite Interactions , Insect Vectors/parasitology , Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/metabolism , Leishmaniasis, Visceral/transmission , Mannose Receptor/metabolism , Mice, Inbred C57BL , Mice, Knockout , Models, Theoretical , Myeloid Cells/immunology , Myeloid Cells/metabolism , Phlebotomus/parasitology , Skin/immunology , Skin/metabolism
4.
Hum Genet ; 139(6-7): 833, 2020 06.
Article in English | MEDLINE | ID: mdl-32445039

ABSTRACT

In the original article publication, the affiliation of the author Ana Coutinho is incorrect.

5.
J Immunol ; 204(11): 2949-2960, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32321759

ABSTRACT

Despite extensive mapping of long noncoding RNAs in immune cells, their function in vivo remains poorly understood. In this study, we identify over 100 long noncoding RNAs that are differentially expressed within 24 h of Th1 cell activation. Among those, we show that suppression of Malat1 is a hallmark of CD4+ T cell activation, but its complete deletion results in more potent immune responses to infection. This is because Malat1-/- Th1 and Th2 cells express lower levels of the immunosuppressive cytokine IL-10. In vivo, the reduced CD4+ T cell IL-10 expression in Malat1-/- mice underpins enhanced immunity and pathogen clearance in experimental visceral leishmaniasis (Leishmania donovani) but more severe disease in a model of malaria (Plasmodium chabaudi chabaudi AS). Mechanistically, Malat1 regulates IL-10 through enhancing expression of Maf, a key transcriptional regulator of IL-10 Maf expression correlates with Malat1 in single Ag-specific Th cells from P. chabaudi chabaudi AS-infected mice and is downregulated in Malat1-/- Th1 and Th2 cells. The Malat1 RNA is responsible for these effects, as antisense oligonucleotide-mediated inhibition of Malat1 also suppresses Maf and IL-10 levels. Our results reveal that through promoting expression of the Maf/IL-10 axis in effector Th cells, Malat1 is a nonredundant regulator of mammalian immunity.


Subject(s)
Interleukin-10/metabolism , Leishmania donovani/physiology , Leishmaniasis, Visceral/immunology , Proto-Oncogene Proteins c-maf/metabolism , RNA, Long Noncoding/genetics , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Female , Gene Expression Regulation , Humans , Immune Tolerance , Immunity/genetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-maf/genetics , Up-Regulation
6.
Cell Host Microbe ; 27(5): 752-768.e7, 2020 05 13.
Article in English | MEDLINE | ID: mdl-32298657

ABSTRACT

The impact of T helper (Th) 1 versus Th2 immunity on intracellular infections is attributed to classical versus alternative activation of macrophages leading to resistance or susceptibility. However, observations in multiple infectious settings demonstrate deficiencies in mediators of Th1-Th2 immunity, which have paradoxical or no impact. We report that prior to influencing activation, Th1/Th2 immunity first controls the size of the permissive host cell reservoir. During early Leishmania infection of the skin, IFN-γ- or STAT6-mediated changes in phagocyte activation were counteracted by changes in IFN-γ-mediated recruitment of permissive CCR2+ monocytes. Monocytes were required for early parasite expansion and acquired an alternatively activated phenotype despite the Th1 dermal environment required for their recruitment. Surprisingly, STAT6 did not enhance intracellular parasite proliferation, but rather modulated the size and permissiveness of the monocytic host cell reservoir via regulation of IFN-γ and IL-10. These observations expand our understanding of the Th1-Th2 paradigm during infection.


Subject(s)
Leishmaniasis/immunology , Monocytes/immunology , Skin/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Female , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interleukin-10/deficiency , Interleukin-10/genetics , Macrophages/immunology , Mice , Mice, Inbred C57BL/genetics , Mice, Knockout , Permissiveness , Psychodidae , Receptors, CCR2/deficiency , Receptors, CCR2/genetics , STAT6 Transcription Factor/deficiency , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism , Virus Replication
7.
Hum Genet ; 139(6-7): 821-831, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32277285

ABSTRACT

Schistosomes induce severe hepatic disease, which is fatal in 2-10% of cases, mortality being higher in cases of co-infection with HBV or HCV. Hepatic disease occurs as a consequence of the chronic inflammation caused by schistosome eggs trapped in liver sinusoids. In certain individuals, the repair process leads to a massive accumulation of fibrosis in the periportal spaces. We and others have shown that genetic variants play a crucial role in disease progression from mild to severe fibrosis and explain why hepatic fibrosis progresses rapidly in certain subjects only. We will review here published findings concerning the strategies that have been used in the analysis of hepatic fibrosis in schistosome-infected individuals, the genetic variants that have associated with fibrosis, and variants in new pathways crucial for fibrosis progression. Together, these studies show that the development of fibrosis is under the tight genetic control of various common variants with moderate effects. This polygenic control has made it possible to develop models that identify schistosome-infected individual at risk of severe hepatic disease. We discuss the performances and limitations of these models.


Subject(s)
Algorithms , Genetic Markers , Liver Diseases, Parasitic/diagnosis , Precision Medicine , Schistosoma/genetics , Schistosomiasis/complications , Severity of Illness Index , Animals , Disease Progression , Humans , Liver Diseases, Parasitic/etiology , Liver Diseases, Parasitic/genetics , Schistosoma/immunology , Schistosoma/pathogenicity , Schistosomiasis/immunology , Schistosomiasis/parasitology
8.
PLoS Pathog ; 16(4): e1008456, 2020 04.
Article in English | MEDLINE | ID: mdl-32282850

ABSTRACT

Leishmania donovani causes visceral leishmaniasis (VL), which is typically fatal without treatment. There is substantial variation between individuals in rates of disease progression, response to treatment and incidence of post-treatment sequelae, specifically post-kala-azar dermal leishmaniasis (PKDL). Nevertheless, the majority of infected people are asymptomatic carriers. Hamsters and mice are commonly used as models of fatal and non-fatal VL, respectively. Host and parasite genetics are likely to be important factors, but in general the reasons for heterogeneous disease presentation in humans and animal models are poorly understood. Host microbiota has become established as a factor in cutaneous forms of leishmaniasis but this has not been studied in VL. We induced intestinal dysbiosis in mice and hamsters by long-term treatment with broad-spectrum antibiotics in their drinking water. There were no significant differences in disease presentation in dysbiotic mice. In contrast, dysbiotic hamsters infected with L. donovani had delayed onset and progression of weight loss. Half of control hamsters had a rapid progression phenotype compared with none of the ABX-treated animals and the nine-month survival rate was significantly improved compared to untreated controls (40% vs. 10%). Antibiotic-treated hamsters also had significantly less severe hepatosplenomegaly, which was accompanied by a distinct cytokine gene expression profile. The protective effect was not explained by differences in parasite loads or haematological profiles. We further found evidence that the gut-liver axis is a key aspect of fatal VL progression in hamsters, including intestinal parasitism, bacterial translocation to the liver, malakoplakia and iron sequestration, none of which occurred in non-progressing murine VL. Diverse bacterial genera were cultured from VL affected livers, of which Rodentibacter was specifically absent from ABX-treated hamsters, indicating this pathobiont may play a role in promoting disease progression. The results provide experimental support for antibiotic prophylaxis against secondary bacterial infections as an adjunct therapy in human VL patients.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Bacterial Infections/prevention & control , Coinfection/prevention & control , Intestinal Diseases, Parasitic/parasitology , Leishmaniasis, Visceral/parasitology , Animals , Antibiotic Prophylaxis , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacterial Physiological Phenomena , Coinfection/microbiology , Cricetinae , Disease Progression , Female , Gastrointestinal Microbiome , Humans , Leishmania donovani/physiology , Leishmaniasis, Visceral/complications , Male , Mesocricetus , Mice , Mice, Inbred C57BL , Symbiosis
9.
PLoS Genet ; 15(5): e1008042, 2019 05.
Article in English | MEDLINE | ID: mdl-31091230

ABSTRACT

Hybrid genotypes have been repeatedly described among natural isolates of Leishmania, and the recovery of experimental hybrids from sand flies co-infected with different strains or species of Leishmania has formally demonstrated that members of the genus possess the machinery for genetic exchange. As neither gamete stages nor cell fusion events have been directly observed during parasite development in the vector, we have relied on a classical genetic analysis to determine if Leishmania has a true sexual cycle. Here, we used whole genome sequencing to follow the chromosomal inheritance patterns of experimental hybrids generated within and between different strains of L. major and L. infantum. We also generated and sequenced the first experimental hybrids in L. tropica. We found that in each case the parental somy and allele contributions matched the inheritance patterns expected under meiosis 97-99% of the time. The hybrids were equivalent to F1 progeny, heterozygous throughout most of the genome for the markers that were homozygous and different between the parents. Rare, non-Mendelian patterns of chromosomal inheritance were observed, including a gain or loss of somy, and loss of heterozygosity, that likely arose during meiosis or during mitotic divisions of the progeny clones in the fly or culture. While the interspecies hybrids appeared to be sterile, the intraspecies hybrids were able to produce backcross and outcross progeny. Analysis of 5 backcross and outcross progeny clones generated from an L. major F1 hybrid, as well as 17 progeny clones generated from backcrosses involving a natural hybrid of L. tropica, revealed genome wide patterns of recombination, demonstrating that classical crossing over occurs at meiosis, and allowed us to construct the first physical and genetic maps in Leishmania. Altogether, the findings provide strong evidence for meiosis-like sexual recombination in Leishmania, presenting clear opportunities for forward genetic analysis and positional cloning of important genes.


Subject(s)
Genome, Protozoan , Leishmania infantum/genetics , Leishmania major/genetics , Leishmania tropica/genetics , Animals , Base Sequence , Chimera , Chromosome Mapping , Crosses, Genetic , Genotype , Inheritance Patterns , Insect Vectors/parasitology , Leishmania infantum/metabolism , Leishmania major/metabolism , Leishmania tropica/metabolism , Meiosis , Psychodidae/parasitology , Recombination, Genetic , Whole Genome Sequencing
10.
Biomed Res Int ; 2018: 3419565, 2018.
Article in English | MEDLINE | ID: mdl-30009168

ABSTRACT

Heren, we analyzed Treg cells as potential biomarkers of disease activity in systemic lupus erythematosus (SLE) patients. Peripheral blood mononuclear cells from 30 SLE patients (15 active: SLEDAI > 6/15 SLE remission: SLEDAI< 6) and 15 healthy volunteers were purified. Treg immunophenotyping was performed using CD4, CD25, CD45, CD127, and FOXP3 markers. CD4+FOXP3+ Treg activation state was investigated based on CD45RA and FOXP3 expression. To increase the accuracy of our findings, a multivariate linear regression was performed. We showed a significant increase in the frequency of CD4+FOXP3+ Treg cells in SLE patients. However, unlike all other Treg cells phenotypes analyzed, only eTreg (CD4+FOXP3highCD45RA-) (p=0.01) subtype was inversely correlated with disease activity while Foxp3+nontreg (CD4+FOXP3lowCD45RA-) (p=0.003) exerted a direct influence in the outcome of the disease. Foxp3+nontreg cells were the most consistent SLE active indicator, confirmed by multiple linear regression analyses. In summary, our results demonstrate Foxp3+nontreg cells as new biomarkers in the search of an effective therapeutic strategy in SLE.


Subject(s)
Biomarkers , Lupus Erythematosus, Systemic/immunology , T-Lymphocytes, Regulatory , Adult , Brazil , CD4 Antigens , Female , Flow Cytometry , Forkhead Transcription Factors , Humans , Interleukin-2 Receptor alpha Subunit , Leukocyte Common Antigens , Leukocytes, Mononuclear , Male , Middle Aged , Young Adult
11.
FASEB J ; 32(3): 1315-1327, 2018 03.
Article in English | MEDLINE | ID: mdl-29097502

ABSTRACT

Leishmania major is the causative agent of the neglected tropical disease, cutaneous leishmaniasis. In the mouse, protective immunity to Leishmania is associated with inflammatory responses. Here, we assess the dynamics of the inflammatory responses at the lesion site during experimental long-term, low-dose intradermal infection of the ear, employing noninvasive imaging and genetically modified L. major. Significant infiltrates of neutrophils and monocytes occurred at 1-4 d and 2-4 wk, whereas dermal macrophage and dendritic cell (DC) numbers were only slightly elevated in the first days. Quantitative whole-body bioluminescence imaging of myeloperoxidase activity and the quantification of parasite loads indicated that the Leishmania virulence factor, inhibitor of serine peptidase 2 (ISP2), is required to modulate phagocyte activation and is important for parasite survival at the infection site. ISP2 played a role in the control of monocyte, monocyte-derived macrophage, and monocyte-derived DC (moDC) influx, and was required to reduce iNOS expression in monocytes, monocyte-derived cells, and dermal DCs; the expression of CD80 in moDCs; and levels of IFN-γ in situ. Our findings indicate that the increased survival of L. major in the dermis during acute infection is associated with the down-regulation of inflammatory monocytes and monocyte-derived cells via ISP2.-Goundry, A., Romano, A., Lima, A. P. C. A., Mottram, J. C., Myburgh, E. Inhibitor of serine peptidase 2 enhances Leishmania major survival in the skin through control of monocytes and monocyte-derived cells.


Subject(s)
Dendritic Cells/immunology , Leishmania major/growth & development , Leishmaniasis, Cutaneous/parasitology , Monocytes/immunology , Serine Endopeptidases/metabolism , Skin/parasitology , Virulence Factors/metabolism , Animals , Cells, Cultured , Female , Leishmania major/immunology , Leishmaniasis, Cutaneous/immunology , Mice , Mice, Inbred C57BL , Skin/immunology
12.
J Exp Med ; 215(1): 357-375, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29247046

ABSTRACT

The origin and functional specialization of dermal macrophages in cutaneous infections have been little studied. In this paper, we show that a strain of Leishmania major (L. major Seidman [LmSd]) that produces nonhealing cutaneous lesions in conventionally resistant C57BL/6 mice was more efficiently taken up by M2-polarized bone marrow (BM)-derived macrophages (BMDMs) in vitro and by mannose receptor (MR)hi dermal macrophages in vivo compared with a healing strain (L. major Friedlin V1). Both in steady and in T helper type 1 (Th1) cell-driven inflammatory states, the MRhi dermal macrophages showed M2 characteristics. The dermal macrophages were radio resistant and not replaced by monocytes or adult BM-derived cells during infection, but were locally maintained by IL-4 and IL-10. Notably, the favored infection of M2 BMDMs by LmSd in vitro was MR dependent, and genetic deletion of MR or selective depletion of MRhi dermal macrophages by anti-CSF-1 receptor antibody reversed the nonhealing phenotype. We conclude that embryonic-derived, MRhi dermal macrophages are permissive for parasite growth even in a strong Th1-immune environment, and the preferential infection of these cells plays a crucial role in the severity of cutaneous disease.


Subject(s)
Lectins, C-Type/metabolism , Leishmania major/immunology , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/metabolism , Macrophages/immunology , Mannose-Binding Lectins/metabolism , Receptors, Cell Surface/metabolism , Th1 Cells/immunology , Animals , Inflammation/immunology , Inflammation/metabolism , Inflammation/virology , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-4/immunology , Interleukin-4/metabolism , Lectins, C-Type/immunology , Leishmaniasis, Cutaneous/virology , Macrophages/metabolism , Mannose Receptor , Mannose-Binding Lectins/immunology , Mice , Mice, Inbred C57BL , Receptors, Cell Surface/immunology , Th1 Cells/metabolism , Th1 Cells/virology
13.
PLoS Pathog ; 13(6): e1006479, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28666021

ABSTRACT

Inflammatory monocytes can be manipulated by environmental cues to perform multiple functions. To define the role of monocytes during primary or secondary infection with an intra-phagosomal pathogen we employed Leishmania major-red fluorescent protein (RFP) parasites and multi-color flow cytometry to define and enumerate infected and uninfected inflammatory cells in the skin. During primary infection, infected monocytes had altered maturation and were the initial mononuclear host cell for parasite replication. In contrast, at a distal site of secondary infection in mice with a healed but persistent primary infection, this same population rapidly produced inducible nitric oxide synthase (iNOS) in an IFN-γ dependent manner and was critical for parasite killing. Maturation to a dendritic cell-like phenotype was not required for monocyte iNOS-production, and enhanced monocyte recruitment correlated with IFN-γ dependent cxcl10 expression. In contrast, neutrophils appeared to be a safe haven for parasites in both primary and secondary sites. Thus, inflammatory monocytes play divergent roles during primary versus secondary infection with an intra-phagosomal pathogen.


Subject(s)
Coinfection/microbiology , Leishmania major , Leishmaniasis, Cutaneous/immunology , Monocytes/microbiology , Phagosomes/metabolism , Skin/microbiology , Animals , Antigens, Ly/immunology , Coinfection/immunology , Dendritic Cells/metabolism , Female , Inflammation/microbiology , Leishmaniasis, Cutaneous/parasitology , Mice, Transgenic , Monocytes/metabolism , Neutrophils/metabolism , Nitric Oxide Synthase Type II/metabolism , Phagosomes/immunology , Receptors, CCR2/immunology , Receptors, Interleukin-8A/immunology
14.
Nat Commun ; 8(1): 57, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28680146

ABSTRACT

Increasing evidence suggests that the infectiousness of patients for the sand fly vector of visceral leishmaniasis is linked to parasites found in the skin. Using a murine model that supports extensive skin infection with Leishmania donovani, spatial analyses at macro-(quantitative PCR) and micro-(confocal microscopy) scales indicate that parasite distribution is markedly skewed. Mathematical models accounting for this heterogeneity demonstrate that while a patchy distribution reduces the expected number of sand flies acquiring parasites, it increases the infection load for sand flies feeding on a patch, increasing their potential for onward transmission. Models representing patchiness at both macro- and micro-scales provide the best fit with experimental sand fly feeding data, pointing to the importance of the skin parasite landscape as a predictor of host infectiousness. Our analysis highlights the skin as a critical site to consider when assessing treatment efficacy, transmission competence and the impact of visceral leishmaniasis elimination campaigns.Parasitemia has been considered the main determinant of visceral leishmaniasis transmission. By combining imaging, qPCR and experimental xenodiagnoses with mathematical models, Doehl et al. argue that the patchy landscape of parasites in the skin is necessary to explain infectiousness.


Subject(s)
Insect Vectors/parasitology , Leishmania donovani/physiology , Leishmaniasis, Visceral/parasitology , Leishmaniasis, Visceral/transmission , Psychodidae/parasitology , Skin/parasitology , Animals , Humans , Mice , Models, Biological , Parasitemia
15.
J Infect Dis ; 215(8): 1285-1293, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28329329

ABSTRACT

Background: Patients with active visceral leishmaniasis are important reservoirs in the anthroponotic transmission cycle of Leishmania donovani. The role of the blood or skin as a source of infection to sand flies remains unclear, and the possible effect of multiple exposures to fly bites on transmissibility has not been addressed. Methods: L. donovani-infected hamsters underwent xenodiagnoses with Lutzomyia longipalpis on the same or different sites on the abdomen on 2 consecutive days or by artificial feeding on the skin or blood. Results: The transmission of L. donovani from sick hamsters to flies was surprisingly low (mean, 24% of fed flies). New flies fed on the same site acquired significantly more infections (mean, 61%; P < .0001). By artificial feeding, flies could acquire infection from blood and skin. However, only artificial feeding on blood produced infections that correlated with the natural feeding (R = 0.792; P < .0001). Infections acquired from blood increased dramatically for blood obtained after exposure to bites, as did the parasitemia level and the number of monocytes in the circulation. Conclusions: The bites of uninfected sand flies favor the transmissibility of L. donovani by infected hosts, owing to a systemic effect that exposure to bites has on the parasitemia. Patients with active visceral leishmaniasis are important reservoirs in the anthroponotic transmission cycle of Leishmania donovani. Using the hamster model of visceral disease, we demonstrate that prior exposure to bites of uninfected sand flies potentiates their ability to transmit infection to the vector.


Subject(s)
Insect Bites and Stings/parasitology , Leishmaniasis, Visceral/transmission , Psychodidae/parasitology , Skin/parasitology , Animals , Cricetinae/parasitology , Female , Insect Vectors/parasitology , Leishmania donovani , Leukocyte Count , Male , Parasite Load , Saliva/parasitology
17.
PLoS Negl Trop Dis ; 10(1): e0004306, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26731721

ABSTRACT

Schistosoma eggs cause chronic liver inflammation and a complex disease characterized by hepatic fibrosis (HF) and splenomegaly (SplM). FOXP3+ Tregs could regulate inflammation, but it is unclear where these cells are produced and what roles they play in human schistosomiasis. We investigated blood and spleen FOXP3+ Tregs in Chinese fishermen with lifelong exposure to Schistosoma japonicum and various degrees of liver and spleen disease. FOXP3+ Tregs accounted for 4.3% of CD4+ T cells and 41.2% of FOXP3+CD4+ T cells; they could be divided into CD45RA-FOXP3hi effector (eTregs) and CD45RA+FOXP3low naive Tregs. Blood Treg levels were high in severe HF (+1.3; p = 0.004) and in SplM (+1.03, p = 0.03). Multivariate regression showed that severe HF (+0.85, p = 0.01) and SplM (+0.97; p = 0.05) were independently associated with the higher proportion of Tregs in the blood. This effect was mostly due to an increase in the proportion of eTregs in the blood of HF+++ (+0.9%; p = 0.04) and SplM (+0.9%; p = 0.04) patients. The proportion of eTregs expressing CXCR3 in the blood was lower in the HF+++ patients (37.4 +/- 5.9%) than in those with milder fibrosis (51.7 ± 2%; p = 0.009), whereas proportion were similar for cells expressing CD25hi, CCR7, and CTLA-4. Splenectomy improves symptoms and was associated with decreases in blood FOXP3+ Treg (-2.5; p<0.001) and eTreg (-1.3; p = 0.03) levels. SplM spleens contained a high proportion of eTregs with CXCR3, CCR5 and CTLA4 upregulation and CCR7 downregulation. This, and the strong expression of ligands of CXCR3 and CCR5 in the liver (n = 8) but not in the spleen suggested that spleen eTregs migrated to Th1-infiltrated liver tissues. Such migration may be attenuated in hepatosplenic patients due to lower levels of CXCR3 expression on Tregs (p = 0.009). Thus, higher blood Treg levels are associated with severe liver disease and splenomegaly. Our data are consistent with the hypothesis that the spleen is a major source of Tregs in subjects with splenomegaly. In most cases, Tregs migrate to the Th1-infiltrated liver and the lower levels of CXCR3+ Tregs in the blood of patients with severe schistosomiasis suggest that decreases in Treg migration sites of inflammation may aggravate the disease.


Subject(s)
Forkhead Transcription Factors/analysis , Liver Cirrhosis/pathology , Schistosomiasis japonica/pathology , Splenomegaly/pathology , T-Lymphocyte Subsets/chemistry , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Aged , Animals , Antigens, CD/analysis , China , Cohort Studies , Humans , Immunophenotyping , Liver/pathology , Liver Cirrhosis/complications , Male , Middle Aged , Occupational Exposure , Spleen/pathology , Splenomegaly/complications , T-Lymphocytes, Regulatory/chemistry
18.
Eur J Immunol ; 46(4): 897-911, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26689285

ABSTRACT

Infection of C57BL/6 mice with most Leishmania major strains results in a healing lesion and clearance of parasites from the skin. Infection of C57BL/6 mice with the L. major Seidman strain (LmSd), isolated from a patient with chronic lesions, despite eliciting a strong Th1 response, results in a nonhealing lesion, poor parasite clearance, and complete destruction of the ear dermis. We show here that in comparison to a healing strain, LmSd elicited early upregulation of IL-1ß mRNA and IL-1ß-producing dermal cells and prominent neutrophil recruitment to the infected skin. Mice deficient in Nlrp3, apoptosis-associated speck-like protein containing a caspase recruitment domain, or caspase-1/11, or lacking IL-1ß or IL-1 receptor signaling, developed healing lesions and cleared LmSd from the infection site. Mice resistant to LmSd had a stronger antigen-specific Th1 response. The possibility that IL-1ß might act through neutrophil recruitment to locally suppress immunity was supported by the healing observed in neutropenic Genista mice. Secretion of mature IL-1ß by LmSd-infected macrophages in vitro was dependent on activation of the Nlrp3 inflammasome and caspase-1. These data reveal that Nlrp3 inflammasome-dependent IL-1ß, associated with localized neutrophil recruitment, plays a crucial role in the development of a nonhealing form of cutaneous leishmaniasis in conventionally resistant mice.


Subject(s)
Carrier Proteins/genetics , Interleukin-1beta/genetics , Leishmania major/immunology , Leishmaniasis, Cutaneous/immunology , Neutrophil Infiltration/immunology , Neutrophils/immunology , Animals , Caspase 1/genetics , Caspases/genetics , Caspases, Initiator , Disease Models, Animal , Humans , Interleukin-17/genetics , Leishmania major/isolation & purification , Leishmaniasis, Cutaneous/parasitology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , RNA, Messenger/biosynthesis , Receptors, Cytokine/genetics , Receptors, Interleukin , Receptors, Interleukin-1 Type I/genetics , Th1 Cells/immunology
19.
J Immunol ; 195(8): 3816-27, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26371247

ABSTRACT

Visceral leishmaniasis (VL) is a fatal disease of the internal organs caused by the eukaryotic parasite Leishmania. Control of VL would best be achieved through vaccination. However, this has proven to be difficult partly because the correlates of protective immunity are not fully understood. In contrast, protective immunity against nonfatal cutaneous leishmaniasis (CL) is well defined and mediated by rapidly recruited, IFN-γ-producing Ly6C(+)CD4(+) T cells at the dermal challenge site. Protection against CL is best achieved by prior infection or live vaccination with Leishmania major, termed leishmanization. A long-standing question is whether prior CL or leishmanization can protect against VL. Employing an intradermal challenge model in mice, we report that cutaneous infection with Leishmania major provides heterologous protection against visceral infection with Leishmania infantum. Protection was associated with a robust CD4(+) T cell response at the dermal challenge site and in the viscera. In vivo labeling of circulating cells revealed that increased frequencies of IFN-γ(+)CD4(+) T cells at sites of infection are due to recruitment or retention of cells in the tissue, rather than increased numbers of cells trapped in the vasculature. Shortly after challenge, IFN-γ-producing cells were highly enriched for Ly6C(+)T-bet(+) cells in the viscera. Surprisingly, this heterologous immunity was superior to homologous immunity mediated by prior infection with L. infantum. Our observations demonstrate a common mechanism of protection against different clinical forms of leishmaniasis. The efficacy of leishmanization against VL may warrant the introduction of the practice in VL endemic areas or during outbreaks of disease.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Leishmania infantum/immunology , Leishmania major/immunology , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/prevention & control , Animals , CD4-Positive T-Lymphocytes/pathology , Female , Interferon-gamma/immunology , Leishmaniasis, Visceral/pathology , Mice
20.
J Immunol ; 194(6): 2664-72, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25672756

ABSTRACT

The outcome of Leishmania infections varies substantially, depending on the host and the parasite strain; infection may be asymptomatic or cause mild or severe skin ulcers (cutaneous leishmaniasis [CL]), limited or disseminated lesions, or lethal visceral disease. We previously reported an association between IL-2R mutations and susceptibility to visceral leishmaniasis in children infected with Leishmania donovani. In the present study, we evaluated the possible role of IL-2 signaling in human CL. We first showed that the transcripts of several genes of the IL-2 pathway were abundant in skin lesions caused by Leishmania braziliensis. We then carried out a genetic analysis, focusing on major genes of the IL-2 pathway. We used a family-based approach and found that polymorphisms of several genes appeared to be associated with CL in a Brazilian population. Moreover, two polymorphisms of the IL2RA gene were significantly and independently associated with CL. We confirmed this result in a second Brazilian sample (also exposed to L. braziliensis) and in Iranians infected with Leishmania tropica: IL2RA rs10905669 T (Pcombined = 6 × 10(-7)) and IL2RA rs706778 T (Pcombined = 2 × 10(-9)) were associated with greater susceptibility to lesion development. These alleles were also correlated with a poor IFN-γ response and poor FOXP3(+) regulatory T cell activation. Thus, IL-2 plays a crucial role in protection against the cutaneous ulcers caused by Leishmania, and the IL-2 pathway is a potential target for strategies aiming to control Leishmania-related diseases.


Subject(s)
Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2/immunology , Leishmania braziliensis/immunology , Leishmaniasis, Cutaneous/immunology , Polymorphism, Single Nucleotide/immunology , Adolescent , Adult , Child , Female , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Gene Frequency , Genetic Predisposition to Disease/genetics , Genotype , Host-Parasite Interactions/immunology , Humans , Interferon-gamma/immunology , Interleukin-2/genetics , Interleukin-2 Receptor alpha Subunit/genetics , Leishmania braziliensis/physiology , Leishmaniasis, Cutaneous/genetics , Leishmaniasis, Cutaneous/parasitology , Linkage Disequilibrium , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide/genetics , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...