Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Parasit Vectors ; 13(1): 96, 2020 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-32087758

RESUMO

BACKGROUND: Since the introduction of miltefosine (MIL) as first-line therapy in the kala-azar elimination programme in the Indian subcontinent, treatment failure rates have been increasing. Since parasite infectivity and virulence may become altered upon treatment relapse, this laboratory study assessed the phenotypic effects of repeated in vitro and in vivo MIL exposure. METHODS: Syngeneic Leishmania donovani lines either or not exposed to MIL were compared for drug susceptibility, rate of promastigote multiplication and metacyclogenesis, macrophage infectivity and behaviour in the sand fly vector, Lutzomyia longipalpis. RESULTS: Promastigotes of both in vitro and in vivo MIL-selected strains displayed a slightly reduced drug susceptibility that was associated with a reduced MIL-accumulation linked to a lower copy number (disomic state) of chromosome 13 harboring the miltefosine transporter (LdMT) gene. In vitro selected promastigotes showed a lower rate of metacyclogenesis whereas the in vivo derived promastigotes displayed a moderately increased growth rate. Repeated MIL exposure did neither influence the parasite load nor metacyclogenesis in the sand fly vector. CONCLUSIONS: Recurrent in vitro and in vivo MIL exposure evokes a number of very subtle phenotypic and genotypic changes which could make promastigotes less susceptible to MIL without attaining full resistance. These changes did not significantly impact on infection in the sand fly vector.


Assuntos
Antiprotozoários/farmacologia , Insetos Vetores/parasitologia , Leishmania donovani/efeitos dos fármacos , Leishmania donovani/fisiologia , Fosforilcolina/análogos & derivados , Psychodidae/parasitologia , Aclimatação , Animais , Resistência a Medicamentos , Humanos , Leishmania donovani/patogenicidade , Leishmaniose Visceral/parasitologia , Leishmaniose Visceral/transmissão , Testes de Sensibilidade Parasitária , Fenótipo , Fosforilcolina/farmacologia , Virulência
2.
Cell Host Microbe ; 23(1): 134-143.e6, 2018 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-29290574

RESUMO

Leishmania donovani parasites are the cause of visceral leishmaniasis and are transmitted by bites from phlebotomine sand flies. A prominent feature of vector-transmitted Leishmania is the persistence of neutrophils at bite sites, where they protect captured parasites, leading to enhanced disease. Here, we demonstrate that gut microbes from the sand fly are egested into host skin alongside Leishmania parasites. The egested microbes trigger the inflammasome, leading to a rapid production of interleukin-1ß (IL-1ß), which sustains neutrophil infiltration. Reducing midgut microbiota by pretreatment of Leishmania-infected sand flies with antibiotics or neutralizing the effect of IL-1ß in bitten mice abrogates neutrophil recruitment. These early events are associated with impairment of parasite visceralization, indicating that both gut microbiota and IL-1ß are important for the establishment of Leishmania infections. Considering that arthropods harbor a rich microbiota, its potential egestion after bites may be a shared mechanism that contributes to severity of vector-borne disease.


Assuntos
Microbioma Gastrointestinal/imunologia , Inflamassomos/imunologia , Interleucina-1beta/imunologia , Leishmania donovani/imunologia , Leishmaniose Visceral/imunologia , Leishmaniose Visceral/transmissão , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Psychodidae/parasitologia , Animais , Antiparasitários/farmacologia , Cricetinae , Feminino , Mordeduras e Picadas de Insetos/parasitologia , Insetos Vetores/parasitologia , Leishmania donovani/efeitos dos fármacos , Leishmaniose Visceral/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Infiltração de Neutrófilos/efeitos dos fármacos , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia
3.
PLoS Pathog ; 13(1): e1006130, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28095465

RESUMO

Differentiation of extracellular Leishmania promastigotes within their sand fly vector, termed metacyclogenesis, is considered to be essential for parasites to regain mammalian host infectivity. Metacyclogenesis is accompanied by changes in the local parasite environment, including secretion of complex glycoconjugates within the promastigote secretory gel and colonization and degradation of the sand fly stomodeal valve. Deletion of the stage-regulated HASP and SHERP genes on chromosome 23 of Leishmania major is known to stall metacyclogenesis in the sand fly but not in in vitro culture. Here, parasite mutants deficient in specific genes within the HASP/SHERP chromosomal region have been used to investigate their role in metacyclogenesis, parasite transmission and establishment of infection. Metacyclogenesis was stalled in HASP/SHERP mutants in vivo and, although still capable of osmotaxis, these mutants failed to secrete promastigote secretory gel, correlating with a lack of parasite accumulation in the thoracic midgut and failure to colonise the stomodeal valve. These defects prevented parasite transmission to a new mammalian host. Sand fly midgut homogenates modulated parasite behaviour in vitro, suggesting a role for molecular interactions between parasite and vector in Leishmania development within the sand fly. For the first time, stage-regulated expression of the small HASPA proteins in Leishmania (Leishmania) has been demonstrated: HASPA2 is expressed only in extracellular promastigotes and HASPA1 only in intracellular amastigotes. Despite its lack of expression in amastigotes, replacement of HASPA2 into the null locus background delays onset of pathology in BALB/c mice. This HASPA2-dependent effect is reversed by HASPA1 gene addition, suggesting that the HASPAs may have a role in host immunomodulation.


Assuntos
Interações Hospedeiro-Parasita/fisiologia , Leishmania major/patogenicidade , Leishmaniose/transmissão , Proteínas de Protozoários/metabolismo , Virulência/fisiologia , Animais , Antígenos de Protozoários/metabolismo , Diferenciação Celular/fisiologia , Modelos Animais de Doenças , Imunofluorescência , Immunoblotting , Insetos Vetores/parasitologia , Leishmania major/crescimento & desenvolvimento , Leishmaniose/genética , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase , Psychodidae/parasitologia
4.
PLoS Negl Trop Dis ; 10(7): e0004771, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27409591

RESUMO

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.


Assuntos
Variação Genética/genética , Leishmania mexicana/fisiologia , Psychodidae/genética , Proteínas e Peptídeos Salivares/metabolismo , Animais , Evolução Molecular , Regulação da Expressão Gênica/fisiologia , Biblioteca Gênica , Psychodidae/parasitologia , Proteínas e Peptídeos Salivares/genética , Transcriptoma
5.
J Infect Dis ; 213(11): 1752-61, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-26768257

RESUMO

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.


Assuntos
Doenças do Cão/parasitologia , Insetos Vetores/parasitologia , Leishmania infantum , Leishmaniose Visceral/veterinária , Psychodidae/parasitologia , Animais , Reservatórios de Doenças/veterinária , Doenças do Cão/imunologia , Doenças do Cão/patologia , Doenças do Cão/transmissão , Cães , Feminino , Humanos , Imunidade Celular , Imunidade Humoral , Mordeduras e Picadas de Insetos/parasitologia , Mordeduras e Picadas de Insetos/patologia , Mordeduras e Picadas de Insetos/veterinária , Interferon gama/metabolismo , Leishmania infantum/isolamento & purificação , Leishmaniose Visceral/parasitologia , Leishmaniose Visceral/patologia , Leishmaniose Visceral/transmissão , Pele/parasitologia , Baço/parasitologia
6.
Cell Rep ; 13(5): 957-67, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26565909

RESUMO

Despite several studies describing the secretion of exosomes by Leishmania in vitro, observation of their formation and release in vivo has remained a major challenge. Herein, we show that Leishmania constitutively secretes exosomes within the lumen of the sand fly midgut through a mechanism homologous to the mammalian pathway. Through egestion experiments, we demonstrate that Leishmania exosomes are part of the sand fly inoculum and are co-egested with the parasite during the insect's bite, possibly influencing the host infectious process. Indeed, co-inoculation of mice footpads with L. major plus midgut-isolated or in-vitro-isolated L. major exosomes resulted in a significant increase in footpad swelling. Notably, co-injections produced exacerbated lesions through overinduction of inflammatory cytokines, in particular IL-17a. Our data indicate that Leishmania exosomes are an integral part of the parasite's infectious life cycle, and we propose to add these vesicles to the repertoire of virulence factors associated with vector-transmitted infections.


Assuntos
Exossomos/metabolismo , Intestinos/parasitologia , Leishmania/patogenicidade , Psychodidae/parasitologia , Animais , Citocinas/metabolismo , Leishmania/metabolismo , Leishmaniose Cutânea/metabolismo , Leishmaniose Cutânea/parasitologia , Leishmaniose Cutânea/patologia , Camundongos
7.
PLoS Negl Trop Dis ; 9(9): e0003991, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26368935

RESUMO

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.


Assuntos
Imunoglobulina G/sangue , Mordeduras e Picadas de Insetos/diagnóstico , Proteínas de Insetos/imunologia , Insetos Vetores , Phlebotomus/imunologia , Proteínas e Peptídeos Salivares/imunologia , Testes Sorológicos/métodos , Adolescente , Animais , Criança , Estudos Transversais , Cães , Feminino , Humanos , Proteínas de Insetos/genética , Masculino , Proteínas e Peptídeos Salivares/genética , Tunísia , Adulto Jovem
8.
Sci Transl Med ; 7(290): 290ra90, 2015 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-26041707

RESUMO

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.


Assuntos
Insetos Vetores , Leishmaniose Cutânea/terapia , Vacinas Protozoárias/uso terapêutico , Psychodidae/parasitologia , Proteínas e Peptídeos Salivares/imunologia , Animais , Humanos , Primatas
9.
FASEB J ; 29(1): 11-24, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25300620

RESUMO

In Leishmania mexicana parasites, a unique glucose transporter, LmxGT1, is selectively targeted to the flagellar membrane, suggesting a possible sensory role that is often associated with ciliary membrane proteins. Expression of LmxGT1 is down-regulated ∼20-fold by increasing cell density but is up-regulated ∼50-fold by depleting glucose from the medium, and the permease is strongly down-regulated when flagellated insect-stage promastigotes invade mammalian macrophages and transform into intracellular amastigotes. Regulation of LmxGT1 expression by glucose and during the lifecycle operates at the level of protein stability. Significantly, a ∆lmxgt1 null mutant, grown in abundant glucose, undergoes catastrophic loss of viability when parasites deplete glucose from the medium, a property not exhibited by wild-type or add-back lines. These results suggest that LmxGT1 may function as a glucose sensor that allows parasites to enter the stationary phase when they deplete glucose and that in the absence of this sensor, parasites do not maintain viability when they run out of glucose. However, alternate roles for LmxGT1 in monitoring glucose availability are considered. The absence of known sensory receptors with defined ligands and biologic functions in Leishmania and related kinetoplastid parasites underscores the potential significance of these observations.


Assuntos
Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Leishmania mexicana/metabolismo , Proteínas de Protozoários/metabolismo , Animais , Linhagem Celular , Feminino , Flagelos/metabolismo , Regulação da Expressão Gênica , Genes de Protozoários , Glucose/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/genética , Humanos , Leishmania mexicana/genética , Leishmania mexicana/patogenicidade , Leishmaniose Cutânea/metabolismo , Leishmaniose Cutânea/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Proteínas de Protozoários/genética , Psychodidae/parasitologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
10.
Front Public Health ; 2: 99, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25152872

RESUMO

Visceral leishmaniasis (VL) is a vector-borne disease transmitted by phlebotomine sand flies and remains the most serious form of the disease with no available human vaccine. Repeatedly, studies have demonstrated the immunogenicity and protective efficacy of a number of sand fly salivary proteins against cutaneous and visceral leishmaniasis. All Leishmania species including agents of VL are co-deposited into the skin together with vector saliva. Generally, the immune response to a protective salivary protein in vaccinated animals is rapid and possibly acts on the parasites soon after delivery into the skin by the bite of an infective sand fly. This is followed by the development of a stronger Leishmania-specific immunity in saliva-vaccinated animals compared to controls. Considering that several of the most efficacious protective molecules were identified from a proven vector of VL, we put forward the notion that a combination vaccine that includes a Leishmania antigen and a vector salivary protein has the potential to improve vaccine efficacy by targeting the parasite at it most vulnerable stage just after transmission.

11.
J Infect Dis ; 207(8): 1328-38, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23288926

RESUMO

BACKGROUND: Visceral leishmaniasis (VL) is transmitted by sand flies. Protection of needle-challenged vaccinated mice was abrogated in vector-initiated cutaneous leishmaniasis, highlighting the importance of developing natural transmission models for VL. METHODS: We used Lutzomyia longipalpis to transmit Leishmania infantum or Leishmania donovani to hamsters. Vector-initiated infections were monitored and compared with intracardiac infections. Body weights were recorded weekly. Organ parasite loads and parasite pick-up by flies were assessed in sick hamsters. RESULTS: Vector-transmitted L. infantum and L. donovani caused ≥5-fold increase in spleen weight compared with uninfected organs and had geometric mean parasite loads (GMPL) comparable to intracardiac inoculation of 10(7)-10(8) parasites, although vector-initiated disease progression was slower and weight loss was greater. Only vector-initiated L. infantum infections caused cutaneous lesions at transmission and distal sites. Importantly, 45.6%, 50.0%, and 33.3% of sand flies feeding on ear, mouth, and testicular lesions, respectively, were parasite-positive. Successful transmission was associated with a high mean percent of metacyclics (66%-82%) rather than total GMPL (2.0 × 10(4)-8.0 × 10(4)) per midgut. CONCLUSIONS: This model provides an improved platform to study initial immune events at the bite site, parasite tropism, and pathogenesis and to test drugs and vaccines against naturally acquired VL.


Assuntos
Modelos Animais de Doenças , Mordeduras e Picadas de Insetos/parasitologia , Insetos Vetores/parasitologia , Leishmaniose Visceral/patologia , Psychodidae/parasitologia , Animais , Peso Corporal , Cricetinae , Progressão da Doença , Leishmania donovani/patogenicidade , Leishmania infantum/patogenicidade , Leishmaniose Cutânea/parasitologia , Leishmaniose Cutânea/patologia , Leishmaniose Cutânea/transmissão , Leishmaniose Visceral/parasitologia , Leishmaniose Visceral/transmissão , Masculino , Tamanho do Órgão , Carga Parasitária , Baço/parasitologia , Baço/patologia
12.
Vet J ; 192(1): 96-100, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21704540

RESUMO

Mycoplasma bovis is a major cause of respiratory outbreaks in cattle feedlots. In this study pulsed-field gel electrophoresis (PFGE) was used to trace field strains and provide information on M. bovis patterns of spread in calf feedlots. The suitability of KpnI, MluI and SmaI restriction enzymes was assessed on different sets of strains. The discriminative power of the first two enzymes was first assessed using 28 epidemiologically unrelated strains; stability was 100% on multiple isolates from in vivo experimental infection. Thirty-nine field isolates from six feedlots were then evaluated. In contrast to the unique fingerprints displayed by the unrelated strains, the isolates from the feedlots showed identical patterns at the time of the outbreak of respiratory disease and 4 weeks later. The PFGE typing results suggest that M. bovis strains follow a clonal epidemic spread pattern at the herd level and that the same strain persists in calves of the herd after the clinical signs have disappeared.


Assuntos
Complexo Respiratório Bovino/epidemiologia , Enzimas de Restrição do DNA/análise , DNA Bacteriano/análise , DNA Bacteriano/genética , Surtos de Doenças/veterinária , Infecções por Mycoplasma/veterinária , Mycoplasma bovis/classificação , Animais , Complexo Respiratório Bovino/microbiologia , Bovinos , Enzimas de Restrição do DNA/genética , Eletroforese em Gel de Campo Pulsado/veterinária , França/epidemiologia , Infecções por Mycoplasma/epidemiologia , Infecções por Mycoplasma/microbiologia , Mycoplasma bovis/genética , Filogenia
13.
J Virol ; 80(15): 7546-54, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16840334

RESUMO

As a preliminary to the localization of the receptor-binding site(s) on the Nipah virus (NiV) glycoprotein (NiV-G), we have undertaken the identification of NiV-G residues that play a role in fusion promotion. To achieve this, we have used two strategies. First, as NiV and Hendra virus (HeV) share a common receptor and their cellular tropism is similar, we hypothesized that residues functioning in receptor attachment could be conserved between their respective G proteins. Our initial strategy was to target charged residues (which can be expected to be at the surface of the protein) conserved between the NiV-G and HeV-G globular heads. Second, we generated NiV variants that escaped neutralization by anti-NiV-G monoclonal antibodies (MAbs) that neutralize NiV both in vitro and in vivo, likely by blocking receptor attachment. The sequencing of such "escape mutants" identified NiV-G residues present in the epitopes to which the neutralizing MAbs are directed. Residues identified via these two strategies whose mutation had an effect on fusion promotion were localized on a new structural model for the NiV-G protein. Our results suggest that seven NiV-G residues, including one (E533) that was identified using both strategies, form a contiguous site on the top of the globular head that is implicated in ephrinB2 binding. This site commences near the shallow depression in the center of the top surface of the globular head and extends to the rim of the barrel-like structure on the top loops of beta-sheet 5. The topology of this site is strikingly similar to that proposed to form the SLAM receptor site on another paramyxovirus attachment protein, that of the measles virus hemagglutinin.


Assuntos
Fusão de Membrana/fisiologia , Modelos Moleculares , Vírus Nipah/fisiologia , Receptores de Superfície Celular/metabolismo , Proteínas do Envelope Viral/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/metabolismo , Sítios de Ligação , Células CHO , Chlorocebus aethiops , Cricetinae , Cricetulus , Efrina-B2/genética , Efrina-B2/metabolismo , Humanos , Dados de Sequência Molecular , Mutação/genética , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Homologia de Sequência de Aminoácidos , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...