Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Phys Act Health ; 21(7): 717-725, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38663845

ABSTRACT

BACKGROUND: To evaluate the influence of previous physical activity (PA) during childhood, adolescence, and current PA practice on the production of antibodies and inflammatory response between the first and second doses of the COVID-19 vaccine. METHODS: Fifty-nine men and 56 women were evaluated before the first vaccine, and 12 weeks later, blood samples were taken to quantify production of anti-severe acute respiratory syndrome coronavirus-2 immunoglobulin G antibodies and cytokines. Previous PA during childhood and adolescence was self-referred, and current PA was assessed using the International Physical Activity Questionnaire. RESULTS: A positive and significant association was observed only between PA practice during adolescence and an increase in antibody production in adulthood (ß = 2012.077, 95% confidence interval, 257.7953-3766.358, P = .025). Individuals who practiced PA during adolescence showed higher production of antibodies between the first and second vaccine dose compared to nonpractitioners (P = .025) and those that accumulated ≥150 minutes per week of current moderate-vigorous PA (MVPA), and presented higher antibody production in relation to who did <150 minutes per week of MVPA (P = .046). Individuals that were practitioners during childhood produced higher G-CSF (P = .047), and those that accumulated ≥150 minutes per week of current MVPA demonstrated lower IP-10 levels (P = .033). However, PA practitioners during adolescence presented higher G-CSF (P = .025), IL-17 (P = .038), IL-1RA (P = .005), IL-1ß (P = .020), and IL-2 (P = .026) levels. CONCLUSION: Our results suggest that adults that accumulated at least 150 minutes of MVPA per week or practiced PA during adolescence developed an improved immune and inflammatory response against COVID-19 vaccination.


Subject(s)
Antibodies, Viral , COVID-19 , Exercise , SARS-CoV-2 , Humans , Male , Female , COVID-19/prevention & control , COVID-19/immunology , Adult , Adolescent , Antibodies, Viral/blood , SARS-CoV-2/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Immunoglobulin G/blood , Immunoglobulin G/immunology , Cytokines/blood , Child , Middle Aged , Young Adult , Inflammation/immunology , Age Factors
2.
Front Immunol ; 15: 1335307, 2024.
Article in English | MEDLINE | ID: mdl-38633260

ABSTRACT

Introduction: Cutaneous leishmaniasis is a neglected vector-borne parasitic disease prevalent in 92 countries with approximately one million new infections annually. Interactions between vector saliva and the human host alter the response to infection and outcome of disease. Methods: To characterize the human immunological responses developed against saliva of Phlebotomus duboscqi, a Leishmania major (L. major) vector, we repeatedly exposed the arms of 14 healthy U.S volunteers to uninfected P. duboscqi bites. Blood was collected a week after each exposure and used to assess total IgG antibodies against the proteins of P. duboscqi salivary gland homogenate (SGH) and the levels of IFN-gamma and IL-10 from peripheral blood mononuclear cells (PBMCs) stimulated with SGH or recombinant sand fly proteins. We analyzed skin punch biopsies of the human volunteer arms from the insect bite site and control skin site after multiple P. duboscqi exposures (four volunteers) using immunohistochemical staining. Results: A variety of immediate insect bite skin reactions were observed. Late skin reactions to insect bites were characterized by macular hyperpigmentation and/or erythematous papules. Hematoxylin and eosin staining showed moderate mononuclear skin infiltrate with eosinophils in those challenged recently (within 2 months), eosinophils were not seen in biopsies with recall challenge (6 month post bites). An increase in plasma antigen-specific IgG responses to SGH was observed over time. Western Blot results showed strong plasma reactivity to five P. duboscqi salivary proteins. Importantly, volunteers developed a cellular immunity characterized by the secretion of IFN-gamma upon PBMC stimulation with P. duboscqi SGH and recombinant antigens. Discussion: Our results demonstrate that humans mounted a local and systemic immune response against P. duboscqi salivary proteins. Specifically, PduM02/SP15-like and PduM73/adenosine deaminase recombinant salivary proteins triggered a Th1 type immune response that might be considered in future development of a potential Leishmania vaccine.


Subject(s)
Insect Bites and Stings , Phlebotomus , Animals , Humans , Phlebotomus/parasitology , Leukocytes, Mononuclear , Immunity, Cellular , Antigens , Immunoglobulin G , Salivary Proteins and Peptides
3.
Mem Inst Oswaldo Cruz ; 119: e230181, 2024.
Article in English | MEDLINE | ID: mdl-38324880

ABSTRACT

BACKGROUND: In Brazil, transmission of visceral and cutaneous leishmaniasis has expanded geographically over the last decades, with both clinical forms occurring simultaneously in the same area. OBJECTIVES: This study characterised the clinical, spatial, and temporal distribution, and performed entomological surveillance and natural infection analysis of a leishmaniasis-endemic area. METHODS: In order to characterise the risk of leishmaniasis transmission in Altos, Piauí, we described the clinical and socio-demographic variables and the spatial and temporal distribution of cases of American visceral leishmaniasis (AVL) and American cutaneous leishmaniasis (ACL) cases and identified potential phlebotomine vectors. FINDINGS: The urban area concentrated almost 54% of ACL and 86.8% of AVL cases. The temporal and spatial distribution of AVL and ACL cases in Altos show a reduction in the number of risk areas, but the presence of permanent disease transmission foci is observed especially in the urban area. 3,808 phlebotomine specimens were captured, with Lutzomyia longipalpis as the most frequent species (98.45%). Of the 35 females assessed for natural infection, one specimen of Lu. longipalpis tested positive for the presence of Leishmania infantum and Leishmania braziliensis DNA. MAIN CONCLUSION: Our results indicate the presence of risk areas for ACL and AVL in the municipality of Altos and highlight the importance of entomological surveillance to further understand a possible role of Lu. longipalpis in ACL transmission.


Subject(s)
Leishmania infantum , Leishmaniasis, Cutaneous , Leishmaniasis, Visceral , Animals , Female , Brazil/epidemiology , Insect Vectors/genetics , Leishmaniasis, Cutaneous/epidemiology , Leishmaniasis, Visceral/epidemiology , Leishmania infantum/genetics , DNA
4.
Mem. Inst. Oswaldo Cruz ; 119: e230181, 2024. tab, graf
Article in English | LILACS-Express | LILACS | ID: biblio-1534931

ABSTRACT

BACKGROUND In Brazil, transmission of visceral and cutaneous leishmaniasis has expanded geographically over the last decades, with both clinical forms occurring simultaneously in the same area. OBJECTIVES This study characterised the clinical, spatial, and temporal distribution, and performed entomological surveillance and natural infection analysis of a leishmaniasis-endemic area. METHODS In order to characterise the risk of leishmaniasis transmission in Altos, Piauí, we described the clinical and socio-demographic variables and the spatial and temporal distribution of cases of American visceral leishmaniasis (AVL) and American cutaneous leishmaniasis (ACL) cases and identified potential phlebotomine vectors. FINDINGS The urban area concentrated almost 54% of ACL and 86.8% of AVL cases. The temporal and spatial distribution of AVL and ACL cases in Altos show a reduction in the number of risk areas, but the presence of permanent disease transmission foci is observed especially in the urban area. 3,808 phlebotomine specimens were captured, with Lutzomyia longipalpis as the most frequent species (98.45%). Of the 35 females assessed for natural infection, one specimen of Lu. longipalpis tested positive for the presence of Leishmania infantum and Leishmania braziliensis DNA. MAIN CONCLUSION Our results indicate the presence of risk areas for ACL and AVL in the municipality of Altos and highlight the importance of entomological surveillance to further understand a possible role of Lu. longipalpis in ACL transmission.

5.
PLoS One ; 18(11): e0287665, 2023.
Article in English | MEDLINE | ID: mdl-38011211

ABSTRACT

Treatment of cutaneous leishmaniasis depends on drugs that potentially cause serious side effects and resistance. Thus, topical therapies are attractive alternatives to the drugs currently used. 3ß, 6ß, 16ß-trihydroxylup-20 (29)-ene is a lupane triterpene isolated from Combretum leprosum Mart. leaves (CLF-1), with reports of in vitro antileishmanial effect against L. amazonensis and to promote lesion healing in animal model. Herein, we evaluated the in vitro and in vivo antileishmanial and healing effects of CLF-1 against L. braziliensis. CLF-1 treatment showed low toxicity in macrophages and significantly reduced parasite load in vitro. CLF-1 induced higher IL-12 and TNF-α production and more discrete IL-4 and IL-10 production. For in vivo evaluation, a CLF-1 cream formulation was prepared to treat hamsters infected with L. braziliensis. CLF-1 treatment was able to reduce parasite load of the infected skin and lymph node more efficiently than the conventional treatment. Histopathological analysis indicated a strong inflammatory response accompanied by an important healing response. Data from this study indicate that topical CLF-1 treatment was effective and non-toxic in L. braziliensis infected hamsters suggesting its potential for further development as a future therapeutic intervention.


Subject(s)
Antiprotozoal Agents , Combretum , Leishmania braziliensis , Leishmaniasis, Cutaneous , Cricetinae , Animals , Mice , Skin/pathology , Leishmaniasis, Cutaneous/drug therapy , Leishmaniasis, Cutaneous/pathology , Wound Healing , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/therapeutic use , Mice, Inbred BALB C
6.
PLoS Negl Trop Dis ; 17(6): e0011388, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37276231

ABSTRACT

BACKGROUND: Leishmaniases are vector borne diseases caused by Leishmania spp. parasites transmitted by female sandflies (Diptera: Psychodidae) whose geographic distribution is influenced by environmental factors. Among the main tools for studying the distribution of vector species, modeling techniques are used to analyze the influence of climatic and environmental factors on the distribution of these insects and their association with human cases of the disease. METHODOLOGY/PRINCIPAL FINDINGS: Here, we used a multiscale ecological niche modeling approach to assess the environmental suitability of sandfly vectors of the etiological agents of Visceral (VL) and American Cutaneous Leishmaniasis (ACL) in Piauí state, northeastern Brazil, and then evaluated their relationship with human disease incidence. For this, we obtained the geographic coordinates of the vector species Lutzomyia longipalpis and Nyssomyia whitmani through literature review, online databases and unpublished records. These data were used for the development of predictive models of the distribution of both sandflies species based on climatic and environmental variables. Finally, the environmental suitability for the presence of these vectors was compared with the incidence of both the diseases at the municipality level. The final models for each sandfly species showed good predictive powers with performance metric values of 0.889 for Lu. longipalpis and 0.776 for Ny. whitmani. The areas with greater environmental suitability for the presence of these species were concentrated in the central-north region of Piauí and coincide with the location of those municipalities presenting higher incidences of VL and ACL, situated in the central-north and extreme north of the state, respectively. The south and southeast regions of Piauí state have low incidence of these diseases and presented low environmental suitability for the presence of both vectors. CONCLUSIONS/SIGNIFICANCE: We discuss how predictive modeling can guide entomological and epidemiological surveillances and recommend an increased supervision and control activities in Teresina (capital of the state of Piaui), Altos and Pedro II, in addition to other municipalities with similar social and environmental characteristics.


Subject(s)
Leishmaniasis, Cutaneous , Phlebotomus , Psychodidae , Animals , Female , Humans , Incidence , Brazil/epidemiology , Insect Vectors/parasitology , Leishmaniasis, Cutaneous/parasitology , Psychodidae/parasitology
7.
Parasite Immunol ; 44(11): e12947, 2022 11.
Article in English | MEDLINE | ID: mdl-36057920

ABSTRACT

The golden hamster (Mesocricetus auratus) is commonly used as a promising model for Leishmania braziliensis infection developing skin-ulcerated lesions. However, different protocols using high concentration of parasites inoculated in the footpad result in severe clinical disease. Here, we further investigate the outcome of the site of infection and concentration of L. braziliensis parasites inoculated on the immunopathogenesis and clinical evolution. Initially, hamsters were infected in the ear dermis or hind footpad with a concentration of 1 × 105 parasites. Animals infected in the ear dermis developed a disease, with an increased parasite load that more closely resembled human cutaneous leishmaniasis lesions comparing to the group infected in the footpad. Next, we evaluated if different parasite concentrations (104 , 105 and 106 ) inoculated in the ear dermis would impact the course and clinical aspects of infection. Hamsters infected with 104 and 105 parasites developed mild lesions compared to the group infected with 106 that presented severe and persistent lesions. The parasite load varied between the different parasite concentrations. The inflammatory response was more intense when infection was initiated with 106 parasites accompanied by an increased initial expression of IL-4, IL-10 and arginase in the lymph node followed by expression of both pro-and anti-inflammatory cytokines comparing to groups infected with 104 and 105 parasites. In conclusion, the number of parasites inoculated, and the initial site of infection could influence the inflammatory response, and clinical presentation. Our results suggest that the ear dermis infection model induces a chronic disease that relates to immunopathological aspects of CL natural infection.


Subject(s)
Leishmania braziliensis , Leishmaniasis, Cutaneous , Animals , Arginase , Cricetinae , Cytokines , Dermis/pathology , Disease Models, Animal , Humans , Interleukin-10 , Interleukin-4 , Leishmaniasis, Cutaneous/parasitology , Mesocricetus
9.
PLoS Negl Trop Dis ; 10(11): e0005078, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27812113

ABSTRACT

BACKGROUND: Previous works showed that immunization with saliva from Lutzomyia intermedia, a vector of Leishmania braziliensis, does not protect against experimental infection. However, L. braziliensis is also transmitted by Lutzomyia whitmani, a sand fly species closely related to Lu. intermedia. Herein we describe the immune response following immunization with Lu. whitmani saliva and the outcome of this response after L. braziliensis infection. METHODS AND FINDINGS: BALB/c mice immunized with Lu. whitmani saliva developed robust humoral and cellular immune responses, the latter characterized by an intense cellular infiltrate and production of IFN-γ and IL-10, by both CD4+ and CD8+ cells. Mice immunized as above and challenged with L. braziliensis plus Lu. whitmani saliva displayed significantly smaller lesions and parasite load at the challenge site. This protection was associated with a higher (p<0.05) IFN-γ production in response to SLA stimulation. Long-term persisting immunity was also detected in mice immunized with Lu. whitmani saliva. Furthermore, individuals residing in an endemic area for cutaneous leishmaniasis (CL) presented antibody responses to Lu. whitmani saliva. However CL patients, with active lesions, displayed a lower humoral response to Lu. whitmani saliva compared to individuals with subclinical Leishmania infection. CONCLUSION: Pre-exposure to Lu. whitmani saliva induces protection against L. braziliensis in a murine model. We also show that Lu. whitmani salivary proteins are immunogenic in naturally exposed individuals. Our results reinforce the importance of investigating the immunomodulatory effect of saliva from different species of closely related sand flies.


Subject(s)
Insect Vectors/immunology , Leishmania braziliensis/immunology , Leishmaniasis, Cutaneous/immunology , Psychodidae/immunology , Saliva/immunology , Salivary Proteins and Peptides/immunology , Animals , Antibodies, Protozoan/blood , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Female , Humans , Immunity, Cellular , Immunity, Humoral , Immunization , Immunoglobulin G/blood , Immunomodulation , Insect Vectors/parasitology , Interferon-gamma/immunology , Interleukin-10/immunology , Leishmaniasis, Cutaneous/parasitology , Leishmaniasis, Cutaneous/prevention & control , Mice , Mice, Inbred BALB C , Parasite Load , Prospective Studies , Psychodidae/parasitology , Saliva/chemistry
10.
J Infect Dis ; 213(11): 1752-61, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26768257

ABSTRACT

Canine leishmaniasis (CanL) is a chronic fatal disease of dogs and a major source of human infection through propagation of parasites in vectors. Here, we infected 8 beagles through multiple experimental vector transmissions with Leishmania infantum-infected Lutzomyia longipalpis. CanL clinical signs varied, although live parasites were recovered from all dog spleens. Splenic parasite burdens correlated positively with Leishmania-specific interleukin 10 levels, negatively with Leishmania-specific interferon γ and interleukin 2 levels, and negatively with Leishmania skin test reactivity. A key finding was parasite persistence for 6 months in lesions observed at the bite sites in all dogs. These recrudesced following a second transmission performed at a distal site. Notably, sand flies efficiently acquired parasites after feeding on lesions at the primary bite site. In this study, controlled vector transmissions identify a potentially unappreciated role for skin at infectious bite sites in dogs with CanL, providing a new perspective regarding the mechanism of Leishmania transmissibility to vector sand flies.


Subject(s)
Dog Diseases/parasitology , Insect Vectors/parasitology , Leishmania infantum , Leishmaniasis, Visceral/veterinary , Psychodidae/parasitology , Animals , Disease Reservoirs/veterinary , Dog Diseases/immunology , Dog Diseases/pathology , Dog Diseases/transmission , Dogs , Female , Humans , Immunity, Cellular , Immunity, Humoral , Insect Bites and Stings/parasitology , Insect Bites and Stings/pathology , Insect Bites and Stings/veterinary , Interferon-gamma/metabolism , Leishmania infantum/isolation & purification , Leishmaniasis, Visceral/parasitology , Leishmaniasis, Visceral/pathology , Leishmaniasis, Visceral/transmission , Skin/parasitology , Spleen/parasitology
11.
Sci Transl Med ; 7(290): 290ra90, 2015 Jun 03.
Article in English | MEDLINE | ID: mdl-26041707

ABSTRACT

Currently, there are no commercially available human vaccines against leishmaniasis. In rodents, cellular immunity to salivary proteins of sand fly vectors is associated to protection against leishmaniasis, making them worthy targets for further exploration as vaccines. We demonstrate that nonhuman primates (NHP) exposed to Phlebotomus duboscqi uninfected sand fly bites or immunized with salivary protein PdSP15 are protected against cutaneous leishmaniasis initiated by infected bites. Uninfected sand fly-exposed and 7 of 10 PdSP15-immunized rhesus macaques displayed a significant reduction in disease and parasite burden compared to controls. Protection correlated to the early appearance of Leishmania-specific CD4(+)IFN-γ(+) lymphocytes, suggesting that immunity to saliva or PdSP15 augments the host immune response to the parasites while maintaining minimal pathology. Notably, the 30% unprotected PdSP15-immunized NHP developed neither immunity to PdSP15 nor an accelerated Leishmania-specific immunity. Sera and peripheral blood mononuclear cells from individuals naturally exposed to P. duboscqi bites recognized PdSP15, demonstrating its immunogenicity in humans. PdSP15 sequence and structure show no homology to mammalian proteins, further demonstrating its potential as a component of a vaccine for human leishmaniasis.


Subject(s)
Insect Vectors , Leishmaniasis, Cutaneous/therapy , Protozoan Vaccines/therapeutic use , Psychodidae/parasitology , Salivary Proteins and Peptides/immunology , Animals , Humans , Primates
12.
J Infect Dis ; 212(1): 157-65, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25596303

ABSTRACT

BACKGROUND: Leishmaniasis is caused by parasites transmitted to the vertebrate host by infected sand flies. During transmission, the vertebrate host is also inoculated with sand fly saliva, which exerts powerful immunomodulatory effects on the host's immune response. METHODS: We conducted a prospective cohort analysis to characterize the human immune response to Lutzomyia intermedia saliva in 264 individuals, from an area for cutaneous leishmaniasis (CL) caused by Leishmania braziliensis. RESULTS: Antibodies were found in 150 individuals (56.8%); immunoglobulin G1 and G4 were the predominant subclasses. Recall responses to salivary gland sonicate showed elevated production of interleukin 10 (IL-10), interleukin 13, interferon γ, CXCL9, and CCL2 compared with controls. CD4(+)CD25(+) T cells, including Foxp3(+) cells, were the main source of IL-10. L. braziliensis replication was increased (P < .05) in macrophages cocultured with saliva-stimulated lymphocytes from exposed individuals and addition of anti-IL-10 reverted this effect. Positive correlation between antibody response to saliva and cellular response to Leishmania was not found. Importantly, individuals seropositive to saliva are 2.1 times more likely to develop CL (relative risk, 2.1; 95% confidence interval, 1.07-4.2; P < .05). CONCLUSIONS: Exposure to L. intermedia sand flies skews the human immune response, facilitating L. braziliensis survival in vitro, and increases the risk of developing CL.


Subject(s)
Disease Susceptibility , Interleukin-10/metabolism , Leishmania braziliensis/isolation & purification , Leishmaniasis, Cutaneous/epidemiology , Leishmaniasis, Cutaneous/immunology , Psychodidae/immunology , Adolescent , Adult , Animals , Antibodies/blood , CD4-Positive T-Lymphocytes/immunology , Child , Child, Preschool , Cohort Studies , Cytokines/blood , Female , Humans , Immunoglobulin G/blood , Male , Middle Aged , Prospective Studies , Risk Assessment , Saliva/immunology , Young Adult
13.
PLoS Negl Trop Dis ; 8(4): e2781, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24762408

ABSTRACT

BACKGROUND: Mice exposed to sand fly saliva are protected against vector-transmitted Leishmania major. Although protection has been related to IFN-γ producing T cells, the early inflammatory response orchestrating this outcome has not been defined. METHODOLOGY/PRINCIPAL FINDINGS: Mice exposed to uninfected P. duboscqi bites and naïve mice were challenged with L. major-infected flies to characterize their early immune response at the bite site. Mostly, chemokine and cytokine transcript expression post-infected bites was amplified in exposed compared to naïve mice. In exposed mice, induced chemokines were mostly involved in leukocyte recruitment and T cell and NK cell activation; IL-4 was expressed at 6 h followed by IFN-γ and iNOS2 as well as IL-5 and IL-10 expression. In naïve animals, the transcript expression following Leishmania-infected sand fly bites was suppressed. Expression profiles translated to an earlier and significantly larger recruitment of leukocytes including neutrophils, macrophages, Gr+ monocytes, NK cells and CD4+ T cells to the bite site of exposed compared to naïve mice post-infected bites. Additionally, up to 48 hours post-infected bites the number of IFN-γ-producing CD4+ T cells and NK cells arriving at the bite site was significantly higher in exposed compared to naïve mice. Thereafter, NK cells become cytolytic and persist at the bite site up to a week post-bite. CONCLUSION/SIGNIFICANCE: The quiet environment induced by a Leishmania-infected sand fly bite in naïve mice was significantly altered in animals previously exposed to saliva of uninfected flies. We propose that the enhanced recruitment of Gr+ monocytes, NK cells and CD4 Th1 cells observed at the bite site of exposed mice creates an inhospitable environment that counters the establishment of L. major infection.


Subject(s)
Bites and Stings/immunology , Bites and Stings/pathology , Leishmania major/immunology , Leishmaniasis, Cutaneous/prevention & control , Psychodidae , Skin/immunology , Skin/pathology , Animals , Cytokines/biosynthesis , Female , Gene Expression , Gene Expression Profiling , Leukocytes/immunology , Mice , Mice, Inbred C57BL
14.
J Invest Dermatol ; 133(2): 452-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22992802

ABSTRACT

Immunity to sand fly saliva in rodents induces a T(H)1 delayed-type hypersensitivity (DTH) response conferring protection against leishmaniasis. The relevance of DTH to sand fly bites in humans living in a leishmaniasis-endemic area remains unknown. Here, we describe the duration and nature of DTH to sand fly saliva in humans from an endemic area of Mali. DTH was assessed at 24, 48, 72, and 96 hours post bite in volunteers exposed to colony-bred sand flies. Dermal biopsies were obtained 48 hours post bite; cytokines were quantified from peripheral blood mononuclear cells (PBMCs) stimulated with sand fly saliva in vitro. A DTH response to bites was observed in 75% of individuals aged 1-15 years, decreasing gradually to 48% by age 45, and dropping to 21% thereafter. Dermal biopsies were dominated by T lymphocytes and macrophages. Abundant expression of IFN-γ and absence of T(H)2 cytokines establishes the T(H)1 nature of this DTH response. PBMCs from 98% of individuals responded to sand fly saliva. Of these, 23% were polarized to a T(H)1 and 25% to a T(H)2 response. We demonstrate the durability and T(H)1 nature of DTH to sand fly bites in humans living in a cutaneous leishmaniasis-endemic area. A systemic T(H)2 response may explain why some individuals remain susceptible to disease.


Subject(s)
Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/parasitology , Leishmania major/immunology , Leishmaniasis/immunology , Psychodidae/immunology , Saliva/immunology , Adolescent , Adult , Aged , Animals , Antigens, Protozoan/immunology , Bites and Stings/immunology , Bites and Stings/parasitology , Child , Disease Susceptibility/epidemiology , Disease Susceptibility/immunology , Endemic Diseases/prevention & control , Endemic Diseases/statistics & numerical data , Female , Humans , Hypersensitivity, Delayed/epidemiology , Leishmaniasis/epidemiology , Leishmaniasis/prevention & control , Male , Mali/epidemiology , Middle Aged , Rodentia , Young Adult
15.
J Immunol ; 189(4): 1535-9, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22798673

ABSTRACT

The environmental factors that contribute to the development of autoimmune diseases are largely unknown. Endemic pemphigus foliaceus in humans, known as Fogo Selvagem (FS) in Brazil, is mediated by pathogenic IgG4 autoantibodies against desmoglein 1 (Dsg1). Clusters of FS overlap with those of leishmaniasis, a disease transmitted by sand fly (Lutzomyia longipalpis) bites. In this study, we show that salivary Ags from the sand fly, and specifically the LJM11 salivary protein, are recognized by FS Abs. Anti-Dsg1 monoclonal autoantibodies derived from FS patients also cross-react with LJM11. Mice immunized with LJM11 generate anti-Dsg1 Abs. Thus, insect bites may deliver salivary Ags that initiate a cross-reactive IgG4 Ab response in genetically susceptible individuals and lead to subsequent FS. Our findings establish a clear relationship between an environmental, noninfectious Ag and the development of potentially pathogenic autoantibodies in an autoimmune disease.


Subject(s)
Autoantibodies/immunology , Cross Reactions , Insect Bites and Stings/complications , Pemphigus/immunology , Psychodidae/immunology , Animals , Antibody Specificity , Autoantigens/immunology , Brazil , Enzyme-Linked Immunosorbent Assay , Humans , Immunoprecipitation , Insect Bites and Stings/epidemiology , Insect Bites and Stings/immunology , Mice , Salivary Glands/immunology
16.
J Invest Dermatol ; 132(12): 2735-43, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22739793

ABSTRACT

Leishmania vaccines that protect against needle challenge fail against the potency of a Leishmania-infected sand fly transmission. Here, we demonstrate that intradermal immunization of mice with 500 ng of the sand fly salivary recombinant protein LJM11 (rLJM11) from Lutzomyia longipalpis, in the absence of adjuvant, induces long-lasting immunity that results in ulcer-free protection against Leishmania major delivered by vector bites. This protection is antibody independent and abrogated by depletion of CD4(+) T cells. Two weeks after challenge, early induction of IFN-γ specifically to rLJM11 correlates to diminished parasite replication in protected animals. At this time point, Leishmania-specific induction of IFN-γ in these mice is low in comparison with its high level in non-protected controls. We hypothesize that early control of parasites in a T-cell helper type 1 environment induced by immunity to LJM11 permits the slow development of Leishmania-specific immunity in the absence of open ulcers. Leishmania-specific immunity observed 5 weeks after infection in rLJM11-immunized mice shows a twofold increase over controls in the percentage of IFN-γ-producing CD4(+) T cells. We propose LJM11 as an immunomodulator that drives an efficient and controlled protective immune response to a sand fly-transmitted Leishmania somewhat mimicking "leishmanization"-induced protective immunity but without its associated lesions.


Subject(s)
Leishmania major/immunology , Leishmaniasis Vaccines/immunology , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/prevention & control , Psychodidae/immunology , Salivary Proteins and Peptides/immunology , Animals , Bites and Stings/immunology , Bites and Stings/parasitology , Disease Models, Animal , Ear, External/immunology , Ear, External/parasitology , HEK293 Cells , Humans , Insect Proteins/immunology , Insect Proteins/pharmacology , Leishmania major/growth & development , Leishmaniasis Vaccines/pharmacology , Leishmaniasis, Cutaneous/transmission , Lymph Nodes/immunology , Lymph Nodes/parasitology , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Psychodidae/parasitology , Saliva/immunology , Saliva/parasitology , Skin Ulcer/immunology , Skin Ulcer/parasitology , Skin Ulcer/prevention & control , Spleen/cytology , Spleen/immunology , Th1 Cells/immunology , Th1 Cells/parasitology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/pharmacology
17.
Front Immunol ; 3: 110, 2012.
Article in English | MEDLINE | ID: mdl-22593758

ABSTRACT

Leishmaniasis is a vector-borne disease transmitted by bites of phlebotomine sand flies. During Leishmania transmission, sand fly saliva is co-inoculated with parasites into the skin of the mammalian host. Sand fly saliva consists of roughly thirty different salivary proteins, many with known roles linked to blood feeding facilitation. Apart from the anti-hemostatic capacity of saliva, several sand fly salivary proteins have been shown to be immunogenic. Immunization with a single salivary protein or exposure to uninfected bites was shown to result in a protective immune response against leishmaniasis. Antibodies to saliva were not required for this protection. A strong body of evidence points to the role for saliva-specific T cells producing IFN-γ in the form of a delayed-type hypersensitivity reaction at the bite site as the main protective response. Herein, we review the immunity to sand fly salivary proteins in the context of its vector-parasite-host combinations and their vaccine potential, as well as some recent advances to shed light on the mechanism of how an immune response to sand fly saliva protects against leishmaniasis.

18.
PLoS Negl Trop Dis ; 6(4): e1610, 2012.
Article in English | MEDLINE | ID: mdl-22509423

ABSTRACT

BACKGROUND: Recombinant KSAC and L110f are promising Leishmania vaccine candidates. Both antigens formulated in stable emulsions (SE) with the natural TLR4 agonist MPL® and L110f with the synthetic TLR4 agonist GLA in SE protected BALB/c mice against L. major infection following needle challenge. Considering the virulence of vector-transmitted Leishmania infections, we vaccinated BALB/c mice with either KSAC+GLA-SE or L110f+GLA-SE to assess protection against L. major transmitted via its vector Phlebotomus duboscqi. METHODS: Mice receiving the KSAC or L110f vaccines were challenged by needle or L. major-infected sand flies. Weekly disease progression and terminal parasite loads were determined. Immunological responses to KSAC, L110f, or soluble Leishmania antigen (SLA) were assessed throughout vaccination, three and twelve weeks after immunization, and one week post-challenge. RESULTS: Following sand fly challenge, KSAC-vaccinated mice were protected while L110f-vaccinated animals showed partial protection. Protection correlated with the ability of SLA to induce IFN-γ-producing CD4(+)CD62L(low)CCR7(low) effector memory T cells pre- and post-sand fly challenge. CONCLUSIONS: This study demonstrates the protective efficacy of KSAC+GLA-SE against sand fly challenge; the importance of vector-transmitted challenge in evaluating vaccine candidates against Leishmania infection; and the necessity of a rapid potent Th1 response against Leishmania to attain true protection.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens, Protozoan/immunology , Leishmania major/immunology , Leishmaniasis Vaccines/immunology , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/prevention & control , Animals , Antibodies, Protozoan/blood , Antigens, Protozoan/administration & dosage , Disease Models, Animal , Female , Interferon-gamma/metabolism , Leishmaniasis Vaccines/administration & dosage , Leishmaniasis, Cutaneous/pathology , Leishmaniasis, Cutaneous/transmission , Mice , Mice, Inbred BALB C , Parasite Load , Phlebotomus/parasitology , T-Lymphocytes/immunology
19.
Arterioscler Thromb Vasc Biol ; 32(3): 786-98, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22116094

ABSTRACT

OBJECTIVE: The coagulation-inflammation cycle has been implicated as a critical component in malaria pathogenesis. Defibrotide (DF), a mixture of DNA aptamers, displays anticoagulant, anti-inflammatory, and endothelial cell (EC)-protective activities and has been successfully used to treat comatose children with veno-occlusive disease. DF was investigated here as a drug to treat cerebral malaria. METHODS AND RESULTS: DF blocks tissue factor expression by ECs incubated with parasitized red blood cells and attenuates prothrombinase activity, platelet aggregation, and complement activation. In contrast, it does not affect nitric oxide bioavailability. We also demonstrated that Plasmodium falciparum glycosylphosphatidylinositol (Pf-GPI) induces tissue factor expression in ECs and cytokine production by dendritic cells. Notably, dendritic cells, known to modulate coagulation and inflammation systemically, were identified as a novel target for DF. Accordingly, DF inhibits Toll-like receptor ligand-dependent dendritic cells activation by a mechanism that is blocked by adenosine receptor antagonist (8-p-sulfophenyltheophylline) but not reproduced by synthetic poly-A, -C, -T, and -G. These results imply that aptameric sequences and adenosine receptor mediate dendritic cells responses to the drug. DF also prevents rosetting formation, red blood cells invasion by P. falciparum and abolishes oocysts development in Anopheles gambiae. In a murine model of cerebral malaria, DF affected parasitemia, decreased IFN-γ levels, and ameliorated clinical score (day 5) with a trend for increased survival. CONCLUSION: Therapeutic use of DF in malaria is proposed.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Anticoagulants/pharmacology , Antimalarials/pharmacology , Blood Coagulation/drug effects , Endothelial Cells/drug effects , Malaria, Cerebral/drug therapy , Plasmodium berghei/drug effects , Plasmodium falciparum/drug effects , Polydeoxyribonucleotides/pharmacology , Animals , Cells, Cultured , Complement Activation/drug effects , Cytokines/blood , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/parasitology , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/parasitology , Female , Glycosylphosphatidylinositols/metabolism , Hemoglobins/metabolism , Humans , Inflammation Mediators/blood , Malaria, Cerebral/blood , Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nitric Oxide/metabolism , Plasmodium berghei/pathogenicity , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Plasmodium falciparum/pathogenicity , Platelet Aggregation/drug effects , Receptors, Purinergic P1/drug effects , Receptors, Purinergic P1/metabolism , Severity of Illness Index , Thromboplastin/metabolism , Time Factors
20.
J Biol Chem ; 286(37): 32383-93, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21795673

ABSTRACT

LJM11, an abundant salivary protein from the sand fly Lutzomyia longipalpis, belongs to the insect "yellow" family of proteins. In this study, we immunized mice with 17 plasmids encoding L. longiplapis salivary proteins and demonstrated that LJM11 confers protective immunity against Leishmania major infection. This protection correlates with a strong induction of a delayed type hypersensitivity (DTH) response following exposure to L. longipalpis saliva. Additionally, splenocytes of exposed mice produce IFN-γ upon stimulation with LJM11, demonstrating the systemic induction of Th1 immunity by this protein. In contrast to LJM11, LJM111, another yellow protein from L. longipalpis saliva, does not produce a DTH response in these mice, suggesting that structural or functional features specific to LJM11 are important for the induction of a robust DTH response. To examine these features, we used calorimetric analysis to probe a possible ligand binding function for the salivary yellow proteins. LJM11, LJM111, and LJM17 all acted as high affinity binders of prohemostatic and proinflammatory biogenic amines, particularly serotonin, catecholamines, and histamine. We also determined the crystal structure of LJM11, revealing a six-bladed ß-propeller fold with a single ligand binding pocket located in the central part of the propeller structure on one face of the molecule. A hypothetical model of LJM11 suggests a positive electrostatic potential on the face containing entry to the ligand binding pocket, whereas LJM111 is negative to neutral over its entire surface. This may be the reason for differences in antigenicity between the two proteins.


Subject(s)
Hypersensitivity, Delayed/immunology , Insect Proteins/immunology , Leishmania major/immunology , Leishmaniasis, Cutaneous/immunology , Psychodidae/immunology , Saliva/immunology , Th1 Cells/immunology , Animals , Biogenic Amines/immunology , Female , Hypersensitivity, Delayed/genetics , Inflammation/genetics , Inflammation/immunology , Insect Proteins/genetics , Insect Proteins/pharmacology , Interferon-gamma/genetics , Interferon-gamma/immunology , Leishmaniasis, Cutaneous/genetics , Leishmaniasis, Cutaneous/prevention & control , Mice , Protein Structure, Tertiary , Psychodidae/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...