Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Front Immunol ; 15: 1335307, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38633260

RESUMEN

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.


Asunto(s)
Mordeduras y Picaduras de Insectos , Phlebotomus , Animales , Humanos , Phlebotomus/parasitología , Leucocitos Mononucleares , Inmunidad Celular , Antígenos , Inmunoglobulina G , Proteínas y Péptidos Salivales
2.
PLoS Negl Trop Dis ; 17(4): e0010862, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37043542

RESUMEN

Phlebotomine sand flies are of global significance as important vectors of human disease, transmitting bacterial, viral, and protozoan pathogens, including the kinetoplastid parasites of the genus Leishmania, the causative agents of devastating diseases collectively termed leishmaniasis. More than 40 pathogenic Leishmania species are transmitted to humans by approximately 35 sand fly species in 98 countries with hundreds of millions of people at risk around the world. No approved efficacious vaccine exists for leishmaniasis and available therapeutic drugs are either toxic and/or expensive, or the parasites are becoming resistant to the more recently developed drugs. Therefore, sand fly and/or reservoir control are currently the most effective strategies to break transmission. To better understand the biology of sand flies, including the mechanisms involved in their vectorial capacity, insecticide resistance, and population structures we sequenced the genomes of two geographically widespread and important sand fly vector species: Phlebotomus papatasi, a vector of Leishmania parasites that cause cutaneous leishmaniasis, (distributed in Europe, the Middle East and North Africa) and Lutzomyia longipalpis, a vector of Leishmania parasites that cause visceral leishmaniasis (distributed across Central and South America). We categorized and curated genes involved in processes important to their roles as disease vectors, including chemosensation, blood feeding, circadian rhythm, immunity, and detoxification, as well as mobile genetic elements. We also defined gene orthology and observed micro-synteny among the genomes. Finally, we present the genetic diversity and population structure of these species in their respective geographical areas. These genomes will be a foundation on which to base future efforts to prevent vector-borne transmission of Leishmania parasites.


Asunto(s)
Leishmania , Leishmaniasis Cutánea , Phlebotomus , Psychodidae , Animales , Humanos , Phlebotomus/parasitología , Psychodidae/parasitología , Leishmania/genética , Genómica
3.
Front Cell Infect Microbiol ; 12: 839932, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35281450

RESUMEN

Sand flies are hematophagous insects responsible for the transmission of vector-borne diseases to humans. Prominent among these diseases is Leishmaniasis that affects the skin and mucous surfaces and organs such as liver and spleen. Importantly, the function of blood-sucking arthropods goes beyond merely transporting pathogens. The saliva of vectors of disease contains pharmacologically active components that facilitate blood feeding and often pathogen establishment. Transcriptomic and proteomic studies have enumerated the repertoire of sand fly salivary proteins and their potential use for the control of Leishmaniasis, either as biomarkers of vector exposure or as anti-Leishmania vaccines. However, a group of specific sand fly salivary proteins triggers formation of cross-reactive antibodies that bind the ectodomain of human desmoglein 1, a member of the epidermal desmosomal cadherins. These cross-reactive antibodies are associated with skin autoimmune blistering diseases, such as pemphigus, in certain immunogenetically predisposed individuals. In this review, we focus on two different aspects of sand fly salivary proteins in the context of human disease: The good, which refers to salivary proteins functioning as biomarkers of exposure or as anti-Leishmania vaccines, and the bad, which refers to salivary proteins as environmental triggers of autoimmune skin diseases.


Asunto(s)
Leishmaniasis , Psychodidae , Animales , Autoinmunidad , Leishmaniasis/prevención & control , Proteómica , Proteínas y Péptidos Salivales
4.
Front Immunol ; 13: 1033705, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36591244

RESUMEN

Regulatory T cells (Tregs) are the master regulators of immunity and they have been implicated in different disease states such as infection, autoimmunity and cancer. Since their discovery, many studies have focused on understanding Treg development, differentiation, and function. While there are many players in the generation and function of truly suppressive Tregs, the role of checkpoint pathways in these processes have been studied extensively. In this paper, we systematically review the role of different checkpoint pathways in Treg homeostasis and function. We describe how co-stimulatory and co-inhibitory pathways modulate Treg homeostasis and function and highlight data from mouse and human studies. Multiple checkpoint pathways are being targeted in cancer and autoimmunity; therefore, we share insights from the clinic and discuss the effect of experimental and approved therapeutics on Treg biology.


Asunto(s)
Neoplasias , Linfocitos T Reguladores , Animales , Humanos , Ratones , Homeostasis , Autoinmunidad
5.
JCI Insight ; 5(23)2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33108348

RESUMEN

A possible etiological link between the onset of endemic pemphigus in Tunisia and bites of Phlebotomus papatasi, the vector of zoonotic cutaneous leishmaniasis, has been previously suggested. We hypothesized that the immunodominant P. papatasi salivary protein PpSP32 binds to desmogleins 1 and 3 (Dsg1 and Dsg3), triggering loss of tolerance to these pemphigus target autoantigens. Here, we show using far-Western blot that the recombinant PpSP32 protein (rPpSP32) binds to epidermal proteins with a MW of approximately 170 kDa. Coimmunoprecipitation revealed the interaction of rPpSP32 with either Dsg1 or Dsg3. A specific interaction between PpSP32 and Dsg1 and Dsg3 was further demonstrated by ELISA assays. Finally, mice immunized with rPpSP32 twice per week exhibited significantly increased levels of anti-Dsg1 and -Dsg3 antibodies from day 75 to 120. Such antibodies were specific for Dsg1 and Dsg3 and were not the result of cross-reactivity to PpSP32. In this study, we demonstrated for the first time to our knowledge a specific binding between PpSP32 and Dsg1 and Dsg3, which might underlie the triggering of anti-Dsg antibodies in patients exposed to sand fly bites. We also confirmed the development of specific anti-Dsg1 and -Dsg3 antibodies in vivo after PpSP32 immunization in mice. Collectively, our results provide evidence that environmental factors, such as the exposure to P. papatasi bites, can trigger the development of autoimmune antibodies.


Asunto(s)
Desmogleínas/inmunología , Pénfigo/etiología , Phlebotomus/inmunología , Adulto , Animales , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Bunyaviridae/inmunología , Bunyaviridae/patogenicidad , Infecciones por Bunyaviridae/inmunología , Cadherinas , Desmogleínas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Tolerancia Inmunológica/inmunología , Inmunoglobulina G , Masculino , Ratones , Pénfigo/inmunología , Psychodidae/inmunología , Proteínas Recombinantes , Túnez/epidemiología
6.
Sci Rep ; 10(1): 7990, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32409684

RESUMEN

Individuals exposed to sand fly bites develop humoral and cellular immune responses to sand fly salivary proteins. Moreover, cellular immunity to saliva or distinct salivary proteins protects against leishmaniasis in various animal models. In Tbilisi, Georgia, an endemic area for visceral leishmaniasis (VL), sand flies are abundant for a short period of ≤3 months. Here, we demonstrate that humans and dogs residing in Tbilisi have little immunological memory to saliva of P. kandelakii, the principal vector of VL. Only 30% of humans and 50% of dogs displayed a weak antibody response to saliva after the end of the sand fly season. Likewise, their peripheral blood mononuclear cells mounted a negligible cellular immune response after stimulation with saliva. RNA seq analysis of wild-caught P. kandelakii salivary glands established the presence of a typical salivary repertoire that included proteins commonly found in other sand fly species such as the yellow, SP15 and apyrase protein families. This indicates that the absence of immunity to P. kandelakii saliva in humans and dogs from Tbilisi is probably caused by insufficient exposure to sand fly bites. This absence of immunity to vector saliva will influence the dynamics of VL transmission in Tbilisi and other endemic areas with brief sand fly seasons.


Asunto(s)
Vectores de Enfermedades , Leishmaniasis/inmunología , Psychodidae/inmunología , Saliva/inmunología , Estaciones del Año , Secuencia de Aminoácidos , Animales , Biomarcadores , Citocinas/metabolismo , Perros , Georgia (República) , Interacciones Huésped-Parásitos/inmunología , Humanos , Mediadores de Inflamación/metabolismo , Leishmaniasis/parasitología , Leishmaniasis/transmisión , Filogenia , Psychodidae/clasificación , Psychodidae/genética , Proteínas y Péptidos Salivales/genética , Proteínas y Péptidos Salivales/inmunología
7.
Clin Immunol ; 207: 49-54, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31325629

RESUMEN

Allergy is a major public health concern, the main treatment for which is symptomatic relief with anti-inflammatory drugs. A key clinical challenge is to induce specific tolerance in order to control allergen-specific memory B and T cells, and specifically block effector cell responses. Our lab recently developed antigen-specific regulatory T-cell (Treg) therapies as a treatment for adverse responses. Recently, we created a chimeric antigen receptor (CAR) approach in which we engineered a target protein antigen, ovalbumin (OVA), linked with the transmembrane and signal transduction domains, CD28-CD3ζ to directly target B cells and sensitized mast cells in an allergy model. We named this receptor "BAR" for B-cell Antibody Receptor. Murine or human Tregs, transduced with a BAR containing OVA or control Tregs expressing an unrelated antigen, were successfully expanded in vitro and tested in the murine OVA-alum allergy model with measurable titers of anti-OVA IgE. Because BAR Tregs express the target antigen and could interact with specific IgE on sensitized mast cells, we first demonstrated that intravenously injected OVA-BAR Tregs did not directly lead to a drop in temperature or release of mediators in plasma indicative of anaphylaxis. Forty-eight hours later, mice were challenged intraperitoneally with 200 µg OVA to induce an anaphylactic reaction, and temperature immediately measured for 30 min. We found that OVA-BAR Tregs protected mice from hypothermia, whereas mice given control BARs (expressing an unrelated antigen) or PBS showed substantial temperature drops indicative of anaphylaxis when systemically challenged with OVA. Importantly, this effect was also demonstrated in a passive anaphylaxis model in which mice that received anti-OVA IgE antibody were protected from hypothermia when treated with OVA-BAR Tregs prior to systemic OVA challenge. These results provide proof of principle that engineered allergen-specific T-regulatory cells can provide clinical protection against severe allergic reactions in individuals already IgE-sensitized to an allergen.


Asunto(s)
Anafilaxia/prevención & control , Ovalbúmina/inmunología , Linfocitos T Reguladores/metabolismo , Alérgenos/inmunología , Animales , Femenino , Tolerancia Inmunológica/inmunología , Inmunización Pasiva , Masculino , Ratones , Ratones Endogámicos BALB C
8.
J Invest Dermatol ; 138(3): 598-606, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29054598

RESUMEN

Nowadays, there is no available vaccine for human leishmaniasis. Animal experiments demonstrate that pre-exposure to sand fly saliva confers protection against leishmaniasis. Our preceding work in humans indicates that Phlebotomus papatasi saliva induces the production of IL-10 by CD8+ T lymphocytes. The neutralization of IL-10 enhanced the activation of a T-cell CD4+ population-producing IFN-γ. Herein, we used a biochemical and functional genomics approach to identify the sand fly salivary components that are responsible for the activation of the T helper type 1 immune response in humans, therefore constituting potential vaccine candidates against leishmaniasis. Fractionated P. papatasi salivary extracts were first tested on T lymphocytes of immune donors. We confirmed that the CD4+ lymphocytes proliferate and produce IFN-γ in response to stimulation with the proteins of molecular weight >30 kDa. Peripheral blood mononuclear cells from immune donors were transfected with plasmids coding for the most abundant proteins from the P. papatasi salivary gland cDNA library. Our result showed that the "yellow related proteins," PPTSP42 and PPTSP44, and "apyrase," PPTSP36, are the proteins responsible for the aforementioned cellular immune response and IFN-γ production. Strikingly, PPTSP44 triggered the highest level of lymphocyte proliferation and IFN-γ production. Multiplex cytokine analysis confirmed the T helper type 1-polarized response induced by these proteins. Importantly, recombinant PPTSP44 validated the results observed with the DNA plasmid, further supporting that PPTSP44 constitutes a promising vaccine candidate against human leishmaniasis.


Asunto(s)
Apirasa/inmunología , Leishmaniasis Cutánea/prevención & control , Phlebotomus/inmunología , Vacunas Antiprotozoos/inmunología , Proteínas y Péptidos Salivales/inmunología , Vacunación , Adolescente , Adulto , Animales , Linfocitos T CD8-positivos/inmunología , Femenino , Humanos , Interferón gamma/biosíntesis , Activación de Linfocitos , Masculino , Células TH1/inmunología , Adulto Joven
9.
Transl Res ; 187: 44-52, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28651073

RESUMEN

Hemophilia A is a bleeding disorder caused by mutations in the gene encoding factor VIII (FVIII), a cofactor protein that is essential for normal blood clotting. Approximately, 1 in 3 patients with severe hemophilia A produce neutralizing antibodies (inhibitors) that block its biologic function in the clotting cascade. Current efforts to eliminate inhibitors consist of repeated FVIII injections under what is termed an "ITI" protocol (Immune Tolerance Induction). However, this method is extremely costly and approximately 30% of patients undergoing ITI do not achieve peripheral tolerance. Human T regulatory cells (Tregs) have been proposed as a new strategy to treat this antidrug antibody response, as well as other diseases. Polyclonal Tregs are nonspecific and could potentially cause general immunosuppression. Novel approaches to induce tolerance to FVIII include the use of engineered human and mouse antigen-specific Tregs, or alternatively antigen-specific cytotoxic cells, to delete, anergize, or kill FVIII-specific lymphocytes. In this review, we discuss the current state of engineered T-cell therapies, and we describe the recent progress in applying these therapies to induce FVIII-specific tolerance.


Asunto(s)
Ingeniería Celular/métodos , Factor VIII/uso terapéutico , Hemofilia A/terapia , Inmunoterapia Adoptiva/métodos , Linfocitos T/fisiología , Factor VIII/genética , Factor VIII/metabolismo , Humanos
10.
PLoS Negl Trop Dis ; 11(3): e0005374, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28278244

RESUMEN

BACKGROUND: Immunity to the sand fly salivary protein SALO (Salivary Anticomplement of Lutzomyia longipalpis) protected hamsters against Leishmania infantum and L. braziliensis infection and, more recently, a vaccine combination of a genetically modified Leishmania with SALO conferred strong protection against L. donovani infection. Because of the importance of SALO as a potential component of a leishmaniasis vaccine, a plan to produce this recombinant protein for future scale manufacturing as well as knowledge of its structural characteristics are needed to move SALO forward for the clinical path. METHODOLOGY/PRINCIPAL FINDINGS: Recombinant SALO was expressed as a soluble secreted protein using Pichia pastoris, rSALO(P), with yields of 1g/L and >99% purity as assessed by SEC-MALS and SDS-PAGE. Unlike its native counterpart, rSALO(P) does not inhibit the classical pathway of complement; however, antibodies to rSALO(P) inhibit the anti-complement activity of sand fly salivary gland homogenate. Immunization with rSALO(P) produces a delayed type hypersensitivity response in C57BL/6 mice, suggesting rSALO(P) lacked anti-complement activity but retained its immunogenicity. The structure of rSALO(P) was solved by S-SAD at Cu-Kalpha to 1.94 Å and refined to Rfactor 17%. SALO is ~80% helical, has no appreciable structural similarities to any human protein, and has limited structural similarity in the C-terminus to members of insect odorant binding proteins. SALO has three predicted human CD4+ T cell epitopes on surface exposed helices. CONCLUSIONS/SIGNIFICANCE: The results indicate that SALO as expressed and purified from P. pastoris is suitable for further scale-up, manufacturing, and testing. SALO has a novel structure, is not similar to any human proteins, is immunogenic in rodents, and does not have the anti-complement activity observed in the native salivary protein which are all important attributes to move this vaccine candidate forward to the clinical path.


Asunto(s)
Psychodidae/química , Proteínas Recombinantes/inmunología , Proteínas y Péptidos Salivales/inmunología , Animales , Expresión Génica , Ratones Endogámicos C57BL , Pichia/genética , Pichia/metabolismo , Conformación Proteica , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas y Péptidos Salivales/administración & dosificación , Proteínas y Péptidos Salivales/química , Proteínas y Péptidos Salivales/genética
11.
PLoS Negl Trop Dis ; 10(7): e0004771, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27409591

RESUMEN

BACKGROUND: Sand fly saliva has been shown to have proteins with potent biological activities, salivary proteins that can be used as biomarkers of vector exposure, and salivary proteins that are candidate vaccines against different forms of leishmaniasis. Sand fly salivary gland transcriptomic approach has contributed significantly to the identification and characterization of many of these salivary proteins from important Leishmania vectors; however, sand fly vectors in some regions of the world are still neglected, as Bichromomyia olmeca (formerly known as Lutzomyia olmeca olmeca), a proven vector of Leishmania mexicana in Mexico and Central America. Despite the importance of this vector in transmitting Leishmania parasite in Mesoamerica there is no information on the repertoire of B. olmeca salivary proteins and their relationship to salivary proteins from other sand fly species. METHODS AND FINDINGS: A cDNA library of the salivary glands of wild-caught B. olmeca was constructed, sequenced, and analyzed. We identified transcripts encoding for novel salivary proteins from this sand fly species and performed a comparative analysis between B. olmeca salivary proteins and those from other sand fly species. With this new information we present an updated catalog of the salivary proteins specific to New World sand flies and salivary proteins common to all sand fly species. We also report in this work the anti-Factor Xa activity of Lofaxin, a salivary anticoagulant protein present in this sand fly species. CONCLUSIONS: This study provides information on the first transcriptome of a sand fly from Mesoamerica and adds information to the limited repertoire of salivary transcriptomes from the Americas. This comparative analysis also shows a fast degree of evolution in salivary proteins from New World sand flies as compared with Old World sand flies.


Asunto(s)
Variación Genética/genética , Leishmania mexicana/fisiología , Psychodidae/genética , Proteínas y Péptidos Salivales/metabolismo , Animales , Evolución Molecular , Regulación de la Expresión Génica/fisiología , Biblioteca de Genes , Psychodidae/parasitología , Proteínas y Péptidos Salivales/genética , Transcriptoma
12.
J Immunol ; 196(5): 2041-50, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26826247

RESUMEN

The etiology of human autoimmune diseases in general remains largely unknown, although the genetic and environmental interplay may be relevant. This applies to the autoimmune diseases of the skin such as the pemphigus phenotypes and others. In this group, there is an endemic form of pemphigus foliaceus (also known as fogo selvagem [FS]) in which the pathogenic IgG4 autoantibody response to the self-antigen desmoglein 1 (Dsg1) cross-reacts with the LJM11 sand fly salivary gland Ag. In this investigation, we dissected the IgG4 autoantibody repertoires used by FS patients in response to endogenous self-Dsg1 and exogenous LJM11 sand fly Ag. Based on analyses of the genetic clonal signatures of these Abs, our results indicate that there is a significant overlap between these two responses, as all identified IgG4 mAbs cross-react to both Dsg1 and LJM11 Ags. Germline H- and L-chain V gene Abs generated according to mutated cross-reactive mAbs preserved their reactivity to both Ags. Our findings suggest that both Dsg1 autoantigen and LJM11 environmental Ag could be the initial antigenic stimulants for the IgG4 autoimmune responses in FS. These results support our hypothesis that LJM11 Ag plays a substantial role in triggering the IgG4 autoantibody development in FS and provide new insights on how noninfectious environmental Ag(s) may drive the generation of autoantibodies in IgG4-related autoimmune diseases.


Asunto(s)
Autoantígenos/inmunología , Inmunoglobulina G/inmunología , Proteínas de Insectos/inmunología , Pénfigo/inmunología , Animales , Autoanticuerpos/inmunología , Reacciones Cruzadas , Desmogleína 1/inmunología , Enfermedades Endémicas , Ensayo de Inmunoadsorción Enzimática , Humanos , Psychodidae/inmunología
13.
PLoS Negl Trop Dis ; 10(1): e0004322, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26752686

RESUMEN

BACKGROUND: Visceral leishmaniasis (VL) is a neglected tropical disease and is fatal if untreated. There is no vaccine available against leishmaniasis. The majority of patients with cutaneous leishmaniasis (CL) or VL develop a long-term protective immunity after cure from infection, which indicates that development of an effective vaccine against leishmaniasis is possible. Such protection may also be achieved by immunization with live attenuated parasites that do not cause disease. We have previously reported a protective response in mice, hamsters and dogs with Leishmania donovani centrin gene knock-out parasites (LdCen-/-), a live attenuated parasite with a cell division specific centrin1 gene deletion. In this study we have explored the effects of salivary protein LJM19 as an adjuvant and intradermal (ID) route of immunization on the efficacy of LdCen-/- parasites as a vaccine against virulent L. donovani. METHODOLOGY/PRINCIPAL FINDINGS: To explore the potential of a combination of LdCen-/- parasites and salivary protein LJM19 as vaccine antigens, LdCen-/- ID immunization followed by ID challenge with virulent L. donovani were performed in hamsters in a 9-month follow up study. We determined parasite burden (serial dilution), antibody production (ELISA) and cytokine expression (qPCR) in these animals. Compared to controls, animals immunized with LdCen-/- + LJM19 induced a strong antibody response, a reduction in spleen and liver parasite burden and a higher expression of pro-inflammatory cytokines after immunization and one month post-challenge. Additionally, a low parasite load in lymph nodes, spleen and liver, and a non-inflamed spleen was observed in immunized animals 9 months after the challenge infection. CONCLUSIONS: Our results demonstrate that an ID vaccination using LdCen-/-parasites in combination with sand fly salivary protein LJM19 has the capability to confer long lasting protection against visceral leishmaniasis that is comparable to intravenous or intracardial immunization.


Asunto(s)
Leishmania donovani/genética , Leishmaniasis Visceral/prevención & control , Proteínas Protozoarias/metabolismo , Vacunas Antiprotozoos/inmunología , Psychodidae/metabolismo , Proteínas y Péptidos Salivales/inmunología , Animales , Cricetinae , Eliminación de Gen , Inyecciones Intradérmicas , Leishmania donovani/inmunología , Proteínas Protozoarias/genética
14.
Sci Rep ; 6: 19300, 2016 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-26758086

RESUMEN

Blood-feeding insects inject potent salivary components including complement inhibitors into their host's skin to acquire a blood meal. Sand fly saliva was shown to inhibit the classical pathway of complement; however, the molecular identity of the inhibitor remains unknown. Here, we identified SALO as the classical pathway complement inhibitor. SALO, an 11 kDa protein, has no homology to proteins of any other organism apart from New World sand flies. rSALO anti-complement activity has the same chromatographic properties as the Lu. longipalpis salivary gland homogenate (SGH)counterparts and anti-rSALO antibodies blocked the classical pathway complement activity of rSALO and SGH. Both rSALO and SGH inhibited C4b deposition and cleavage of C4. rSALO, however, did not inhibit the protease activity of C1s nor the enzymatic activity of factor Xa, uPA, thrombin, kallikrein, trypsin and plasmin. Importantly, rSALO did not inhibit the alternative or the lectin pathway of complement. In conclusion our data shows that SALO is a specific classical pathway complement inhibitor present in the saliva of Lu. longipalpis. Importantly, due to its small size and specificity, SALO may offer a therapeutic alternative for complement classical pathway-mediated pathogenic effects in human diseases.


Asunto(s)
Inactivadores del Complemento/farmacología , Vía Clásica del Complemento/efectos de los fármacos , Proteínas de Insectos/farmacología , Psychodidae/inmunología , Psychodidae/metabolismo , Saliva/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Activación de Complemento/efectos de los fármacos , Complemento C1/antagonistas & inhibidores , Complemento C1/inmunología , Complemento C1/metabolismo , Complemento C4/antagonistas & inhibidores , Complemento C4/inmunología , Complemento C4/metabolismo , Humanos , Proteínas Recombinantes/farmacología
16.
PLoS Negl Trop Dis ; 9(9): e0003991, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26368935

RESUMEN

BACKGROUND: During a blood meal, female sand flies, vectors of Leishmania parasites, inject saliva into the host skin. Sand fly saliva is composed of a large variety of components that exert different pharmacological activities facilitating the acquisition of blood by the insect. Importantly, proteins present in saliva are able to elicit the production of specific anti-saliva antibodies, which can be used as markers for exposure to vector bites. Serological tests using total sand fly salivary gland extracts are challenging due to the difficulty of obtaining reproducible salivary gland preparations. Previously, we demonstrated that PpSP32 is the immunodominant salivary antigen in humans exposed to Phlebotomus papatasi bites and established that humans exposed to P. perniciosus bites do not recognize it. METHODOLOGY/PRINCIPAL FINDINGS: Herein, we have validated, in a large cohort of 522 individuals, the use of the Phlebotomus papatasi recombinant salivary protein PpSP32 (rPpSP32) as an alternative method for testing exposure to the bite of this sand fly. We also demonstrated that screening for total anti-rPpSP32 IgG antibodies is sufficient, being comparable in efficacy to the screening for IgG2, IgG4 and IgE antibodies against rPpSP32. Additionally, sera obtained from dogs immunized with saliva of P. perniciosus, a sympatric and widely distributed sand fly in Tunisia, did not recognize rPpSP32 demonstrating its suitability as a marker of exposure to P. papatasi saliva. CONCLUSIONS/SIGNIFICANCE: Our data indicate that rPpSP32 constitutes a useful epidemiological tool to monitor the spatial distribution of P. papatasi in a particular region, to direct control measures against zoonotic cutaneous leishmaniasis, to assess the efficiency of vector control interventions and perhaps to assess the risk of contracting the disease.


Asunto(s)
Inmunoglobulina G/sangre , Mordeduras y Picaduras de Insectos/diagnóstico , Proteínas de Insectos/inmunología , Insectos Vectores , Phlebotomus/inmunología , Proteínas y Péptidos Salivales/inmunología , Pruebas Serológicas/métodos , Adolescente , Animales , Niño , Estudios Transversales , Perros , Femenino , Humanos , Proteínas de Insectos/genética , Masculino , Proteínas y Péptidos Salivales/genética , Túnez , Adulto Joven
18.
Sci Transl Med ; 7(290): 290ra90, 2015 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-26041707

RESUMEN

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.


Asunto(s)
Insectos Vectores , Leishmaniasis Cutánea/terapia , Vacunas Antiprotozoos/uso terapéutico , Psychodidae/parasitología , Proteínas y Péptidos Salivales/inmunología , Animales , Humanos , Primates
19.
PLoS Negl Trop Dis ; 9(2): e0003449, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25646796

RESUMEN

BACKGROUND: The sandfly Phlebotomus papatasi is the vector of Leishmania major, the main causative agent of Old World cutaneous leishmaniasis (CL) in Saudi Arabia. Sandflies inject saliva while feeding and the salivary protein PpSP32 was previously shown to be a biomarker for bite exposure. Here we used recombinant PpSP32 to evaluate human exposure to Ph. papatasi bites, and study the association between antibody response to saliva and CL in endemic areas in Saudi Arabia. METHODOLOGY/PRINCIPAL FINDINGS: In this observational study, anti-PpSP32 antibodies, as indicators of exposure to sandfly bites, were measured in sera from healthy individuals and patients from endemic regions in Saudi Arabia with active and cured CL. Ph. papatasi was identified as the primary CL vector in the study area. Anti-PpSP32 antibody levels were significantly higher in CL patients presenting active infections from all geographical regions compared to CL cured and healthy individuals. Furthermore, higher anti-PpSP32 antibody levels correlated with the prevalence and type of CL lesions (nodular vs. papular) observed in patients, especially non-local construction workers. CONCLUSIONS: Our findings suggest a possible correlation between the type of immunity generated by the exposure to sandfly bites and disease outcome.


Asunto(s)
Anticuerpos Antiprotozoarios/sangre , Mordeduras y Picaduras de Insectos/parasitología , Leishmania major/inmunología , Leishmaniasis Cutánea/patología , Phlebotomus/parasitología , Proteínas y Péptidos Salivales/inmunología , Adolescente , Adulto , Animales , Anticuerpos Antiprotozoarios/inmunología , Formación de Anticuerpos/inmunología , Línea Celular , Femenino , Humanos , Insectos Vectores/parasitología , Leishmaniasis Cutánea/inmunología , Masculino , Persona de Mediana Edad , Pronóstico , Saliva/inmunología , Saliva/metabolismo , Arabia Saudita , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...