Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Mol Ecol ; 31(5): 1444-1460, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34905257

RESUMO

In animals with distinct life stages such as holometabolous insects, adult phenotypic variation is often shaped by the environment of immature stages, including their interactions with microbes colonizing larval habitats. Such carry-over effects were previously observed for several adult traits of the mosquito Aedes aegypti after larval exposure to different bacteria, but the mechanistic underpinnings are unknown. Here, we investigated the molecular changes triggered by gnotobiotic larval exposure to different bacteria in Ae. aegypti. We initially screened a panel of 16 bacterial isolates from natural mosquito breeding sites to determine their ability to influence adult life-history traits. We subsequently focused on four bacterial isolates (belonging to Flavobacterium, Lysobacter, Paenibacillus, and Enterobacteriaceae) with significant carry-over effects on adult survival and found that they were associated with distinct transcriptomic profiles throughout mosquito development. Moreover, we detected carry-over effects at the level of gene expression for the Flavobacterium and Paenibacillus isolates. The most prominent transcriptomic changes in gnotobiotic larvae reflected a profound remodelling of lipid metabolism, which translated into phenotypic differences in lipid storage and starvation resistance at the adult stage. Together, our findings indicate that larval exposure to environmental bacteria trigger substantial physiological changes that impact adult fitness, uncovering a possible mechanism underlying carry-over effects of mosquito-bacteria interactions during larval development.


Assuntos
Aedes , Aedes/microbiologia , Animais , Bactérias/genética , Ecossistema , Larva/microbiologia
2.
PLoS Pathog ; 14(1): e1006794, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29352310

RESUMO

Leishmania parasites are transmitted to vertebrate hosts by female phlebotomine sand flies as they bloodfeed by lacerating the upper capillaries of the dermis with their barbed mouthparts. In the sand fly midgut secreted proteophosphoglycans from Leishmania form a biological plug known as the promastigote secretory gel (PSG), which blocks the gut and facilitates the regurgitation of infective parasites. The interaction between the wound created by the sand fly bite and PSG is not known. Here we nanoinjected a sand fly egested dose of PSG into BALB/c mouse skin that lead to the differential expression of 7,907 transcripts. These transcripts were transiently up-regulated during the first 6 hours post-wound and enriched for pathways involved in inflammation, cell proliferation, fibrosis, epithelial cell differentiation and wound remodelling. We found that PSG significantly accelerated wound healing in vitro and in mice; which was associated with an early up-regulation of transcripts involved in inflammation (IL-1ß, IL-6, IL-10, TNFα) and inflammatory cell recruitment (CCL2, CCL3, CCL4, CXCL2), followed 6 days later by enhanced expression of transcripts associated with epithelial cell proliferation, fibroplasia and fibrosis (FGFR2, EGF, EGFR, IGF1). Dermal expression of IGF1 was enhanced following an infected sand fly bite and was acutely responsive to the deposition of PSG but not the inoculation of parasites or sand fly saliva. Antibody blockade of IGF1 ablated the gel's ability to promote wound closure in mouse ears and significantly reduced the virulence of Leishmania mexicana infection delivered by an individual sand fly bite. Dermal macrophages recruited to air-pouches on the backs of mice revealed that IGF1 was pivotal to the PSG's ability to promote macrophage alternative activation and Leishmania infection. Our data demonstrate that through the regurgitation of PSG Leishmania exploit the wound healing response of the host to the vector bite by promoting the action of IGF1 to drive the alternative activation of macrophages.


Assuntos
Fator de Crescimento Insulin-Like I/fisiologia , Leishmaniose Cutânea/patologia , Proteínas de Membrana/farmacologia , Proteoglicanas/farmacologia , Proteínas de Protozoários/farmacologia , Psychodidae/metabolismo , Pele/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Animais , Células Cultivadas , Progressão da Doença , Feminino , Interações Hospedeiro-Parasita/fisiologia , Leishmania mexicana/metabolismo , Leishmaniose Cutânea/parasitologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/parasitologia , Macrófagos/patologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Proteoglicanas/metabolismo , Proteínas de Protozoários/metabolismo , Transdução de Sinais/efeitos dos fármacos , Pele/parasitologia , Pele/patologia
3.
BMC Microbiol ; 19(1): 32, 2019 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-30736736

RESUMO

BACKGROUND: Leishmania (L.) spp are intracellular eukaryotic parasites responsible for cutaneous or visceral leishmaniasis, replicating predominantly in macrophages (MF). In C57BL/6 mice virulence with L. amazonensis has been associated with inhibition of Th1 immune responses and an uncontrolled lesion development, whereas DBA/2 mice control any lesion. Parasitic clearance by the MFs requires the activation of proper immune responses. One of the immune related genes expressed in immune cells including MF, codes for osteopontin (OPN). OPN is a secreted glycoprotein, acting as an immune regulator. Its implication in promoting Th1 immunity in response to infectious microorganisms and its known protective effect against viral and bacterial infections via activation of the immune response, render OPN a molecule of interest in the study of the host response to L. amazonensis. RESULTS: We examined the host response to L. amazonensis of opn mutant and wild type C57BL/6 mice. Bone marrow derived MFs were infected with the parasites in vitro, and opn mutant and wild type mice were inoculated in vivo by intradermal injection in the ears. The DBA/2 strain known to control L. amazonensis infection was also used for comparison. Our data indicate that the parasites increased opn gene expression and OPN protein while parasitic proliferation was contained in the presence of OPN. In the presence of parasites the expression of inflammation-related transcripts was inhibited. Interleukin-1-beta (IL-1ß), and transcripts of the NLR-family (NLRC4, NLRP3) were down regulated after L. amazonensis infection. In the absence of OPN, the inhibition by the parasites of IL-1ß transcripts was less efficient and a pyroptosis-like cell phenotype was detected in vitro, suggesting a central role of OPN in the host-response to L. amazonensis. Similarly, in vivo, in the absence of OPN, while the clinical inflammatory phenotype is more severe, an increase of these transcripts was observed. CONCLUSIONS: L. amazonensis infection induces opn gene expression and protein, which in turn participates in shaping the host response to the parasites, seemingly by decreasing the activation of inflammation. OPN, further evaluated as a target for Leishmaniasis control represents an additional interest in improving vaccination strategies against the parasites.


Assuntos
Interações Hospedeiro-Parasita/imunologia , Leishmaniose Visceral/imunologia , Macrófagos/imunologia , Osteopontina/imunologia , Animais , Feminino , Inflamação , Interferon gama/genética , Interferon gama/imunologia , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Leishmania braziliensis , Macrófagos/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Osteopontina/genética , Células Th1/imunologia
4.
J Proteome Res ; 14(1): 318-29, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25369177

RESUMO

Parasitic infections such as leishmaniasis induce a cascade of host physiological responses, including metabolic and immunological changes. Infection with Leishmania major parasites causes cutaneous leishmaniasis in humans, a neglected tropical disease that is difficult to manage. To understand the determinants of pathology, we studied L. major infection in two mouse models: the self-healing C57BL/6 strain and the nonhealing BALB/c strain. Metabolic profiling of urine, plasma, and feces via proton NMR spectroscopy was performed to discover parasite-specific imprints on global host metabolism. Plasma cytokine status and fecal microbiome were also characterized as additional metrics of the host response to infection. Results demonstrated differences in glucose and lipid metabolism, distinctive immunological phenotypes, and shifts in microbial composition between the two models. We present a novel approach to integrate such metrics using correlation network analyses, whereby self-healing mice demonstrated an orchestrated interaction between the biological measures shortly after infection. In contrast, the response observed in nonhealing mice was delayed and fragmented. Our study suggests that trans-system communication across host metabolism, the innate immune system, and gut microbiome is key for a successful host response to L. major and provides a new concept, potentially translatable to other diseases.


Assuntos
Biomarcadores/metabolismo , Microbioma Gastrointestinal/imunologia , Leishmania major/imunologia , Leishmaniose Cutânea/imunologia , Leishmaniose Cutânea/fisiopatologia , Modelos Biológicos , Animais , Biomarcadores/sangue , Biomarcadores/urina , Interações Hospedeiro-Patógeno , Leishmaniose Cutânea/metabolismo , Espectroscopia de Ressonância Magnética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Especificidade da Espécie
5.
iScience ; 27(6): 110019, 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38883823

RESUMO

The COVID-19 pandemic highlighted the need for antivirals against emerging coronaviruses (CoV). Inhibiting spike (S) glycoprotein-mediated viral entry is a promising strategy. To identify small molecule inhibitors that block entry downstream of receptor binding, we established a high-throughput screening (HTS) platform based on pseudoviruses. We employed a three-step process to screen nearly 200,000 small molecules. First, we identified hits that inhibit pseudoviruses bearing the SARS-CoV-2 S glycoprotein. Counter-screening against pseudoviruses with the vesicular stomatitis virus glycoprotein (VSV-G), yielded sixty-five SARS-CoV-2 S-specific inhibitors. These were further tested against pseudoviruses bearing the MERS-CoV S glycoprotein, which uses a different receptor. Out of these, five compounds, which included the known broad-spectrum inhibitor Nafamostat, were subjected to further validation and tested against pseudoviruses bearing the S glycoprotein of the Alpha, Delta, and Omicron variants as well as bona fide SARS-CoV-2. This rigorous approach revealed an unreported inhibitor and its derivative as potential broad-spectrum antivirals.

6.
Med Trop Sante Int ; 3(2)2023 06 30.
Artigo em Francês | MEDLINE | ID: mdl-37525683

RESUMO

To attempt resolving this issue accurately, it was necessary to anchor our experimental approaches in the observations and pioneering work of our predecessors, notably Alphonse Laveran, Louis Parrot, Edmond and Étienne Sergent. The latter, among other things, had identified as natural hosts of leishmaniasis, rodent populations with which hematophagous telmophagous sand fly populations cohabited closely.When human populations emerged in these natural ecosystems, after the sedentarization of Homo sapiens, more or less important disturbances would have led to a transition of sand fly hematophagy, from zoophilia, to zoo-anthropophilia and anthropophilia.The creation of infrastructures that allow the breeding and integration into experimental groups of both holobiont sand flies and holobiont laboratory rodents (rats, mice, hamsters, etc.) remains crucial. With such infrastructures, it becomes possible to grasp and characterize the multilateral dynamic processes - mostly clinically silent - that account for the biogenesis of tissue and/or cellular niches protecting populations of Leishmania developmental morphotypes, including those ensuring host-to-host transmission, albeit in small numbers.


Assuntos
Leishmania , Leishmaniose , Phlebotomus , Psychodidae , Cricetinae , Humanos , Camundongos , Ratos , Animais , Ecossistema , Roedores
7.
Cold Spring Harb Protoc ; 2023(7): .pdb.prot108176, 2023 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-36882291

RESUMO

The biteOscope enables the high-resolution monitoring and video recording of blood-feeding mosquitoes. Mosquito biting is induced by combining host cues, an artificial bloodmeal, a membrane, and a transparent heater in a transparent behavioral arena. Machine vision techniques enable the tracking and pose estimation of individual mosquitoes to discern behavior and resolve individual feeding events. The workflow allows multiple replicates and large amounts of imaging data to be generated rapidly. These data are suitable for downstream analysis using machine learning tools for behavioral analysis, allowing subtle behavioral effects to be characterized.


Assuntos
Culicidae , Comportamento Alimentar , Animais
8.
Cold Spring Harb Protoc ; 2023(7): .pdb.top107658, 2023 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-36882292

RESUMO

Blood feeding is a critical event in the life cycle of female mosquitoes. In addition to providing nutrients to the mosquito, blood feeding facilitates the transmission of parasites and viruses to hosts, potentially having devastating health consequences. Our understanding of these short, yet important, bouts of behavior is incomplete. How and where a mosquito decides to bite and the success of feeding can influence the transmission of pathogens. A more thorough understanding of these processes may allow the development of interventions that reduce or prevent infections. Here, we present an overview of strategies for studying mosquito biting behavior and introduce the biteOscope, which provides an opportunity to observe and understand this behavior at unprecedented spatial and temporal resolution under tightly controlled conditions. The biteOscope combines recent advances in computer vision and automated tracking with designs for behavioral arenas and controllable artificial host cues that use low-cost and readily available materials.


Assuntos
Culicidae , Mordeduras e Picadas de Insetos , Animais , Feminino , Comportamento Alimentar
9.
iScience ; 26(4): 106413, 2023 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-36968074

RESUMO

The landscape of SARS-CoV-2 variants dramatically diversified with the simultaneous appearance of multiple subvariants originating from BA.2, BA.4, and BA.5 Omicron sub-lineages. They harbor a specific set of mutations in the spike that can make them more evasive to therapeutic monoclonal antibodies. In this study, we compared the neutralizing potential of monoclonal antibodies against the Omicron BA.2.75.2, BQ.1, BQ.1.1, and XBB variants, with a pre-Omicron Delta variant as a reference. Sotrovimab retains some activity against BA.2.75.2, BQ.1, and XBB as it did against BA.2/BA.5, but is less active against BQ.1.1. Within the Evusheld/AZD7442 cocktail, Cilgavimab lost all activity against all subvariants studied, resulting in loss of Evusheld activity. Finally, Bebtelovimab, while still active against BA.2.75, also lost all neutralizing activity against BQ.1, BQ.1.1, and XBB variants.

10.
Cells ; 11(23)2022 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-36497121

RESUMO

CEP55 is a central regulator of late cytokinesis and is overexpressed in numerous cancers. Its post-translationally controlled recruitment to the midbody is crucial to the structural coordination of the abscission sequence. Our recent evidence that CEP55 contains two ubiquitin-binding domains was the first structural and functional link between ubiquitin signaling and ESCRT-mediated severing of the intercellular bridge. So far, high-content screens focusing on cytokinesis have used multinucleation as the endpoint readout. Here, we report an automated image-based detection method of intercellular bridges, which we applied to further our understanding of late cytokinetic signaling by performing an RNAi screen of ubiquitin ligases and deubiquitinases. A secondary validation confirmed four candidate genes, i.e., LNX2, NEURL, UCHL1 and RNF157, whose downregulation variably affects interconnected phenotypes related to CEP55 and its UBDs, as follows: decreased recruitment of CEP55 to the midbody, increased number of midbody remnants per cell, and increased frequency of intercellular bridges or multinucleation events. This brings into question the Notch-dependent or independent contributions of LNX2 and NEURL proteins to late cytokinesis. Similarly, the role of UCHL1 in autophagy could link its function with the fate of midbody remnants. Beyond the biological interest, this high-content screening approach could also be used to isolate anticancer drugs that act by impairing cytokinesis and CEP55 functions.


Assuntos
Proteínas Nucleares , Ubiquitina , Humanos , Ubiquitina/metabolismo , Proteínas Nucleares/metabolismo , Citocinese/fisiologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Ligação Proteica
11.
J Exp Med ; 219(7)2022 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-35704748

RESUMO

Memory B-cell and antibody responses to the SARS-CoV-2 spike protein contribute to long-term immune protection against severe COVID-19, which can also be prevented by antibody-based interventions. Here, wide SARS-CoV-2 immunoprofiling in Wuhan COVID-19 convalescents combining serological, cellular, and monoclonal antibody explorations revealed humoral immunity coordination. Detailed characterization of a hundred SARS-CoV-2 spike memory B-cell monoclonal antibodies uncovered diversity in their repertoire and antiviral functions. The latter were influenced by the targeted spike region with strong Fc-dependent effectors to the S2 subunit and potent neutralizers to the receptor-binding domain. Amongst those, Cv2.1169 and Cv2.3194 antibodies cross-neutralized SARS-CoV-2 variants of concern, including Omicron BA.1 and BA.2. Cv2.1169, isolated from a mucosa-derived IgA memory B cell demonstrated potency boost as IgA dimers and therapeutic efficacy as IgG antibodies in animal models. Structural data provided mechanistic clues to Cv2.1169 potency and breadth. Thus, potent broadly neutralizing IgA antibodies elicited in mucosal tissues can stem SARS-CoV-2 infection, and Cv2.1169 and Cv2.3194 are prime candidates for COVID-19 prevention and treatment.


Assuntos
COVID-19 , SARS-CoV-2 , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , Humanos , Imunoglobulina A , Imunoglobulina G , Glicoproteína da Espícula de Coronavírus
12.
Sci Rep ; 11(1): 3266, 2021 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-33547379

RESUMO

West Nile virus (WNV) is a Flavivirus, which can cause febrile illness in humans that may progress to encephalitis. Like any other obligate intracellular pathogens, Flaviviruses hijack cellular protein functions as a strategy for sustaining their life cycle. Many cellular proteins display globular domain known as PDZ domain that interacts with PDZ-Binding Motifs (PBM) identified in many viral proteins. Thus, cellular PDZ-containing proteins are common targets during viral infection. The non-structural protein 5 (NS5) from WNV provides both RNA cap methyltransferase and RNA polymerase activities and is involved in viral replication but its interactions with host proteins remain poorly known. In this study, we demonstrate that the C-terminal PBM of WNV NS5 recognizes several human PDZ-containing proteins using both in vitro and in cellulo high-throughput methods. Furthermore, we constructed and assayed in cell culture WNV replicons where the PBM within NS5 was mutated. Our results demonstrate that the PBM of WNV NS5 is important in WNV replication. Moreover, we show that knockdown of the PDZ-containing proteins TJP1, PARD3, ARHGAP21 or SHANK2 results in the decrease of WNV replication in cells. Altogether, our data reveal that interactions between the PBM of NS5 and PDZ-containing proteins affect West Nile virus replication.


Assuntos
Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/fisiologia , Animais , Sítios de Ligação , Linhagem Celular , Células HEK293 , Humanos , Domínios PDZ , Proteínas não Estruturais Virais/química , Febre do Nilo Ocidental/metabolismo
13.
Bio Protoc ; 10(11): e3633, 2020 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-33659304

RESUMO

Parasites of the genus Leishmania infect the mammalian hosts, including mice and humans and cause cutaneous or visceral leishmaniasis depending upon the parasite species transmitted by the vector sandfly. Leishmania amazonensis is one of the Leishmania species responsible for the cutaneous form of the disease. We have inoculated with these parasites the ear dermis of mice. RNA preparations were performed from fragmented tissues using a buffer containing guanidin isothiocynate (RLT buffer, RNeasy Mini Kit, Qiagen, SAS, France) and ß-mercaptoethanol. Both reagents facilitate the isolation of intact RNA from tissues and the use of the RNeasy Kits present with several advantages that facilitate the isolation of pure non-degraded total RNA: i) This method allows to avoid the presence of phenol in the RNA extraction buffer, commonly used in alternative protocols; ii) Moreover Diethylpyrocarbonate (DEPC) treatment of glassware, to avoid RNAses contamination of the samples, is not required with this protocol; iii) Finally, it is a fast procedure and the isolated total RNA may be concentrated in a small volume thus facilitating its use for downstream experimental procedures.

14.
Commun Biol ; 2: 84, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30854476

RESUMO

Predicting how Leishmania will respond to control efforts requires an understanding of their transmission strategy. Using real-time quantitative PCR to quantify infectious metacyclic and non-metacyclic forms in mouse skin from single sandfly bites we show that most transmissions were highly enriched for infectious parasites. However, a quarter of sandflies were capable of transmitting high doses containing more non-infectious promastigotes from the vector's midgut. Mouse infections replicating "high" to "low" quality, low-dose transmissions confirmed clear differences in the pathology of the infection and their onward transmissibility back to sandflies. Borrowing methods originally developed to account for exposure heterogeneity among hosts, we show how these high-dose, low-quality transmitters act as super-spreading vectors, capable of inflating Leishmania transmission potential by as much as six-fold. These results highlight the hidden potential of transmission of mixed Leishmania promastigote stages on disease prevalence and the role of dose heterogeneity as an underlying strategy for efficient transmission.


Assuntos
Mordeduras e Picadas de Insetos , Leishmania , Leishmaniose/parasitologia , Leishmaniose/transmissão , Psychodidae/parasitologia , Animais , Modelos Animais de Doenças , Feminino , Leishmania/crescimento & desenvolvimento , Leishmania/isolamento & purificação , Leishmaniose/diagnóstico , Leishmaniose Cutânea/parasitologia , Camundongos , Carga Parasitária
15.
Parasitol Int ; 63(1): 245-53, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24001683

RESUMO

Leishmania/L. major was identified as the etiological agent of human localized cutaneous leishmaniasis. L. major metacyclic promastigotes/MP - the infectious form transmitted by sand flies - were enriched from axenically-derived cultures and inoculated into the dermis of mice (10(3) or 10(4) luciferase-expressing L. major MP inoculated into the C57BL/6 mouse ear pinna). Quantitative readout assays were then combined with imaging of this L. major-hosting skin site and established i) that a specific period of time - depending upon the L. major load used for the inoculation - is required for the L. major-hosting ear pinna to be continuously populated by a balanced population of functional regulatory and effector T lymphocytes, and that ii) this balance coincides with persisting low numbers of amastigotes in more or less rapidly healing skin. This approach also established that, whatever the MP inoculum load delivered to the primary site, the immune processes that reduce the L. major amastigote population also account for concomitant immunity, namely remodelling of the secondary site - where 10(4) MP were delivered - as a clinically silent niche hosting a small L. major population.


Assuntos
Pavilhão Auricular/parasitologia , Leishmania major/metabolismo , Leishmania major/fisiologia , Luciferases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Animais , Regulação da Expressão Gênica/imunologia , Leishmania major/genética , Luciferases/genética , Medições Luminescentes , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Tempo
16.
PLoS Negl Trop Dis ; 7(6): e2276, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23785538

RESUMO

BACKGROUND: After loading with live Leishmania (L) amazonensis amastigotes, mouse myeloid dendritic leucocytes/DLs are known to undergo reprogramming of their immune functions. In the study reported here, we investigated whether the presence of live L. amazonensis amastigotes in mouse bone marrow-derived DLs is able to trigger re-programming of DL lipid, and particularly neutral lipid metabolism. METHODOLOGY/PRINCIPAL FINDINGS: Affymetrix-based transcriptional profiles were determined in C57BL/6 and DBA/2 mouse bone marrow-derived DLs that had been sorted from cultures exposed or not to live L. amazonensis amastigotes. This showed that live amastigote-hosting DLs exhibited a coordinated increase in: (i) long-chain fatty acids (LCFA) and cholesterol uptake/transport, (ii) LCFA and cholesterol (re)-esterification to triacyl-sn-glycerol (TAG) and cholesteryl esters (CE), respectively. As these neutral lipids are known to make up the lipid body (LB) core, oleic acid was added to DL cultures and LB accumulation was compared in live amastigote-hosting versus amastigote-free DLs by epi-fluorescence and transmission electron microscopy. This showed that LBs were both significantly larger and more numerous in live amastigote-hosting mouse dendritic leucocytes. Moreover, many of the larger LB showed intimate contact with the membrane of the parasitophorous vacuoles hosting the live L. amazonensis amastigotes. CONCLUSIONS/SIGNIFICANCE: As leucocyte LBs are known to be more than simple neutral lipid repositories, we set about addressing two related questions. Could LBs provide lipids to live amastigotes hosted within the DL parasitophorous vacuole and also deliver? Could LBs impact either directly or indirectly on the persistence of L. amazonensis amastigotes in rodent skin?


Assuntos
Células Dendríticas/metabolismo , Células Dendríticas/parasitologia , Interações Hospedeiro-Patógeno , Leishmania mexicana/fisiologia , Metabolismo dos Lipídeos , Animais , Feminino , Perfilação da Expressão Gênica , Leishmania mexicana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA
17.
PLoS Negl Trop Dis ; 6(12): e1980, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23272268

RESUMO

BACKGROUND/OBJECTIVES: The inoculation of a low number (10(4)) of L. amazonensis metacyclic promastigotes into the dermis of C57BL/6 and DBA/2 mouse ear pinna results in distinct outcome as assessed by the parasite load values and ear pinna macroscopic features monitored from days 4 to 22-phase 1 and from days 22 to 80/100-phase 2. While in C57BL/6 mice, the amastigote population size was increasing progressively, in DBA/2 mice, it was rapidly controlled. This latter rapid control did not prevent intracellular amastigotes to persist in the ear pinna and in the ear-draining lymph node/ear-DLN. The objectives of the present analysis was to compare the dendritic leukocytes-dependant immune processes that could account for the distinct outcome during the phase 1, namely, when phagocytic dendritic leucocytes of C57BL/6 and DBA/2 mice have been subverted as live amastigotes-hosting cells. METHODOLOGY/PRINCIPAL FINDINGS: Being aware of the very low frequency of the tissues' dendritic leucocytes/DLs, bone marrow-derived C57BL/6 and DBA/2 DLs were first generated and exposed or not to live DsRed2 expressing L. amazonensis amastigotes. Once sorted from the four bone marrow cultures, the DLs were compared by Affymetrix-based transcriptomic analyses and flow cytometry. C57BL/6 and DBA/2 DLs cells hosting live L. amazonensis amastigotes do display distinct transcriptional signatures and markers that could contribute to the distinct features observed in C57BL/6 versus DBA/2 ear pinna and in the ear pinna-DLNs during the first phase post L. amazonensis inoculation. CONCLUSIONS/SIGNIFICANCE: The distinct features captured in vitro from homogenous populations of C57BL/6 and DBA/2 DLs hosting live amastigotes do offer solid resources for further comparing, in vivo, in biologically sound conditions, functions that range from leukocyte mobilization within the ear pinna, the distinct emigration from the ear pinna to the DLN of live amastigotes-hosting DLs, and their unique signalling functions to either naive or primed T lymphocytes.


Assuntos
Células Dendríticas/parasitologia , Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno , Leishmania mexicana/patogenicidade , Animais , Feminino , Leishmania mexicana/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA