Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Genet ; 11(3): e1005120, 2015 03.
Artigo em Inglês | MEDLINE | ID: mdl-25815810

RESUMO

Anaplasma phagocytophilum is an emerging pathogen that causes human granulocytic anaplasmosis. Infection with this zoonotic pathogen affects cell function in both vertebrate host and the tick vector, Ixodes scapularis. Global tissue-specific response and apoptosis signaling pathways were characterized in I. scapularis nymphs and adult female midguts and salivary glands infected with A. phagocytophilum using a systems biology approach combining transcriptomics and proteomics. Apoptosis was selected for pathway-focused analysis due to its role in bacterial infection of tick cells. The results showed tissue-specific differences in tick response to infection and revealed differentiated regulation of apoptosis pathways. The impact of bacterial infection was more pronounced in tick nymphs and midguts than in salivary glands, probably reflecting bacterial developmental cycle. All apoptosis pathways described in other organisms were identified in I. scapularis, except for the absence of the Perforin ortholog. Functional characterization using RNA interference showed that Porin knockdown significantly increases tick colonization by A. phagocytophilum. Infection with A. phagocytophilum produced complex tissue-specific alterations in transcript and protein levels. In tick nymphs, the results suggested a possible effect of bacterial infection on the inhibition of tick immune response. In tick midguts, the results suggested that A. phagocytophilum infection inhibited cell apoptosis to facilitate and establish infection through up-regulation of the JAK/STAT pathway. Bacterial infection inhibited the intrinsic apoptosis pathway in tick salivary glands by down-regulating Porin expression that resulted in the inhibition of Cytochrome c release as the anti-apoptotic mechanism to facilitate bacterial infection. However, tick salivary glands may promote apoptosis to limit bacterial infection through induction of the extrinsic apoptosis pathway. These dynamic changes in response to A. phagocytophilum in I. scapularis tissue-specific transcriptome and proteome demonstrated the complexity of the tick response to infection and will contribute to characterize gene regulation in ticks.


Assuntos
Anaplasma phagocytophilum/genética , Anaplasmose/genética , Apoptose/genética , Biologia de Sistemas , Anaplasma phagocytophilum/patogenicidade , Anaplasmose/microbiologia , Anaplasmose/transmissão , Animais , Diferenciação Celular/genética , Feminino , Regulação da Expressão Gênica , Humanos , Insetos Vetores/genética , Insetos Vetores/microbiologia , Ixodes/microbiologia , Especificidade de Órgãos , Interferência de RNA , Glândulas Salivares/metabolismo , Glândulas Salivares/microbiologia , Transdução de Sinais/genética , Transcriptoma/genética
2.
Mol Cell Proteomics ; 14(12): 3154-72, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26424601

RESUMO

Anaplasma phagocytophilum is an emerging zoonotic pathogen that causes human granulocytic anaplasmosis. These intracellular bacteria establish infection by affecting cell function in both the vertebrate host and the tick vector, Ixodes scapularis. Previous studies have characterized the tick transcriptome and proteome in response to A. phagocytophilum infection. However, in the postgenomic era, the integration of omics datasets through a systems biology approach allows network-based analyses to describe the complexity and functionality of biological systems such as host-pathogen interactions and the discovery of new targets for prevention and control of infectious diseases. This study reports the first systems biology integration of metabolomics, transcriptomics, and proteomics data to characterize essential metabolic pathways involved in the tick response to A. phagocytophilum infection. The ISE6 tick cells used in this study constitute a model for hemocytes involved in pathogen infection and immune response. The results showed that infection affected protein processing in endoplasmic reticulum and glucose metabolic pathways in tick cells. These results supported tick-Anaplasma co-evolution by providing new evidence of how tick cells limit pathogen infection, while the pathogen benefits from the tick cell response to establish infection. Additionally, ticks benefit from A. phagocytophilum infection by increasing survival while pathogens guarantee transmission. The results suggested that A. phagocytophilum induces protein misfolding to limit the tick cell response and facilitate infection but requires protein degradation to prevent ER stress and cell apoptosis to survive in infected cells. Additionally, A. phagocytophilum may benefit from the tick cell's ability to limit bacterial infection through PEPCK inhibition leading to decreased glucose metabolism, which also results in the inhibition of cell apoptosis that increases infection of tick cells. These results support the use of this experimental approach to systematically identify cell pathways and molecular mechanisms involved in tick-pathogen interactions. Data are available via ProteomeXchange with identifier PXD002181.


Assuntos
Anaplasma phagocytophilum/fisiologia , Ehrlichiose/veterinária , Metabolômica/métodos , Proteômica/métodos , Carrapatos/microbiologia , Animais , Linhagem Celular , Ehrlichiose/genética , Ehrlichiose/metabolismo , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Glucose/metabolismo , Interações Hospedeiro-Patógeno , Redes e Vias Metabólicas , Biologia de Sistemas/métodos
3.
J Gen Virol ; 96(Pt 7): 1676-81, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25701823

RESUMO

In 2011, a neurological disease was reported in a herd of goats (Capra hircus) in Asturias, Spain. Initial sequencing identified the causative agent as louping ill virus (LIV). Subsequently, with the application of whole genome sequencing and phylogenetic analysis, empirical data demonstrates that the LIV-like virus detected is significantly divergent from LIV and Spanish sheep encephalitis virus (SSEV). This virus encoded an amino acid sequence motif at the site of a previously identified marker for differentiating tick-borne flaviviruses that was shared with a virus previously isolated in Ireland in 1968. The significance of these observations reflects the diversity of tick-borne flaviviruses in Europe. These data also contribute to our knowledge of the evolution of tick-borne flaviviruses and could reflect the movement of viruses throughout Europe. Based on these observations, the proposed name for this virus is Spanish goat encephalitis virus (SGEV), to distinguish it from SSEV.


Assuntos
Vírus da Encefalite Transmitidos por Carrapatos/isolamento & purificação , Encefalite Transmitida por Carrapatos/veterinária , Genoma Viral , Doenças das Cabras/virologia , Cabras/virologia , RNA Viral/genética , Análise de Sequência de DNA , Animais , Análise por Conglomerados , Vírus da Encefalite Transmitidos por Carrapatos/genética , Encefalite Transmitida por Carrapatos/virologia , Irlanda , Dados de Sequência Molecular , Filogenia , Homologia de Sequência , Espanha
4.
Infect Immun ; 81(7): 2415-25, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23630955

RESUMO

Anaplasma phagocytophilum causes human granulocytic anaplasmosis. Infection with this zoonotic pathogen affects gene expression in both the vertebrate host and the tick vector, Ixodes scapularis. Here, we identified new genes, including spectrin alpha chain or alpha-fodrin (CG8) and voltage-dependent anion-selective channel or mitochondrial porin (T2), that are involved in A. phagocytophilum infection/multiplication and the tick cell response to infection. The pathogen downregulated the expression of CG8 in tick salivary glands and T2 in both the gut and salivary glands to inhibit apoptosis as a mechanism to subvert host cell defenses and increase infection. In the gut, the tick response to infection through CG8 upregulation was used by the pathogen to increase infection due to the cytoskeleton rearrangement that is required for pathogen infection. These results increase our understanding of the role of tick genes during A. phagocytophilum infection and multiplication and demonstrate that the pathogen uses similar strategies to establish infection in both vertebrate and invertebrate hosts.


Assuntos
Anaplasma phagocytophilum/patogenicidade , Apoptose , Proteínas de Transporte/metabolismo , Citoesqueleto/metabolismo , Ixodes/microbiologia , Proteínas dos Microfilamentos/metabolismo , Anaplasma phagocytophilum/genética , Animais , Proteínas de Transporte/genética , Caspase 9/genética , Caspase 9/metabolismo , Linhagem Celular , Comportamento Alimentar , Feminino , Trato Gastrointestinal/microbiologia , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Interações Hospedeiro-Patógeno , Ixodes/genética , Ixodes/metabolismo , Masculino , Proteínas dos Microfilamentos/genética , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Filogenia , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Glândulas Salivares/microbiologia , Espectrina/genética , Espectrina/metabolismo , Canais de Ânion Dependentes de Voltagem/genética , Canais de Ânion Dependentes de Voltagem/metabolismo
6.
Parasitol Res ; 111(3): 1391-5, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22411632

RESUMO

The horn fly Haematobia irritans (Linnaeus, 1758) (Diptera: Muscidae) is one of the most important ectoparasites of cattle. The parasitism of horn flies interferes with cattle feeding, thus reducing weight gain and milk production. Additionally, horn flies are mechanical vectors of pathogens that cause disease in cattle. The aims of this study were to identify microorganisms in partially fed female horn flies through mining of expressed sequence tags (ESTs) and to characterize microorganism prevalence using real-time RT-PCR. Seven unigenes containing 24 ESTs were homologous to infectious agents. Microorganisms identified in partially fed female horn flies ESTs included Nora virus (3 unigenes; 8 ESTs), Wolbachia endosymbionts (3 unigenes; 3 ESTs), and Mycobacterium bovis (1 unigene; 13 ESTs). These results expanded the repertoire of microorganisms that could cause persistent infections or be mechanically transmitted by horn flies and support further studies on the role of horn flies in the epidemiology of these pathogens in Mexico.


Assuntos
Dípteros/microbiologia , Dípteros/fisiologia , Mycobacterium bovis/isolamento & purificação , Picornaviridae/isolamento & purificação , Wolbachia/isolamento & purificação , Animais , Bovinos/sangue , Etiquetas de Sequências Expressas , Feminino , Mycobacterium bovis/classificação , Mycobacterium bovis/genética , Picornaviridae/classificação , Picornaviridae/genética , DNA Polimerase Dirigida por RNA , Reação em Cadeia da Polimerase em Tempo Real , Wolbachia/classificação , Wolbachia/genética
7.
BMC Genomics ; 12: 105, 2011 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-21310032

RESUMO

BACKGROUND: The horn fly, Haematobia irritans (Linnaeus, 1758) (Diptera: Muscidae) is one of the most important ectoparasites of pastured cattle. Horn flies infestations reduce cattle weight gain and milk production. Additionally, horn flies are mechanical vectors of different pathogens that cause disease in cattle. The aim of this study was to conduct a functional genomics study in female horn flies using Expressed Sequence Tags (EST) analysis and RNA interference (RNAi). RESULTS: A cDNA library was made from whole abdominal tissues collected from partially fed adult female horn flies. High quality horn fly ESTs (2,160) were sequenced and assembled into 992 unigenes (178 contigs and 814 singlets) representing molecular functions such as serine proteases, cell metabolism, mitochondrial function, transcription and translation, transport, chromatin structure, vitellogenesis, cytoskeleton, DNA replication, cell response to stress and infection, cell proliferation and cell-cell interactions, intracellular trafficking and secretion, and development. Functional analyses were conducted using RNAi for the first time in horn flies. Gene knockdown by RNAi resulted in higher horn fly mortality (protease inhibitor functional group), reduced oviposition (vitellogenin, ferritin and vATPase groups) or both (immune response and 5'-NUC groups) when compared to controls. Silencing of ubiquitination ESTs did not affect horn fly mortality and oviposition while gene knockdown in the ferritin and vATPse functional groups reduced mortality when compared to controls. CONCLUSIONS: These results advanced the molecular characterization of this important ectoparasite and suggested candidate protective antigens for the development of vaccines for the control of horn fly infestations.


Assuntos
Genoma de Inseto , Genômica , Muscidae/genética , Animais , Etiquetas de Sequências Expressas , Feminino , Biblioteca Gênica , Interferência de RNA , Análise de Sequência de DNA
8.
Artigo em Inglês | MEDLINE | ID: mdl-28491823

RESUMO

Campylobacter jejuni is the leading food-borne poisoning in industrialized countries. While the bacteria causes disease in humans, it merely colonizes the gut in poultry or pigs, where seems to establish a commensal relationship. Until now, few studies have been conducted to elucidate the relationship between C. jejuni and its different hosts. In this work, a comparative proteomics approach was used to identify the underlying mechanisms involved in the divergent outcome following C. jejuni infection in human and porcine host. Human (INT-407) and porcine (IPEC-1) intestinal cell lines were infected by C. jejuni for 3 h (T3h) and 24 h (T24h). C. jejuni infection prompted an intense inflammatory response at T3h in human intestinal cells, mainly characterized by expression of proteins involved in cell spreading, cell migration and promotion of reactive oxygen species (ROS). Proteomic analysis evidenced significantly regulated biofunctions in human cells related with engulfment and endocytosis, and supported by canonical pathways associated to infection such as caveolar- and clathrin-mediated endocytosis signaling. In porcine IPEC-1 cells, inflammatory response as well as signaling pathways that control cellular functions such as cell migration, endocytosis and cell cycle progression resulted downregulated. These differences in the host response to infection were supported by the different pattern of adhesion and invasion proteins expressed by C. jejuni in human and porcine cells. No marked differences in expression of virulence factors involved in adaptive response and iron acquisition functions were observed. Therefore, the results of this study suggest that both host and pathogen factors are responsible for commensal or infectious character of C. jejuni in different hosts.


Assuntos
Infecções por Campylobacter/microbiologia , Campylobacter jejuni/patogenicidade , Interações Hospedeiro-Patógeno/fisiologia , Proteômica/métodos , Simbiose/fisiologia , Animais , Proteínas de Bactérias/metabolismo , Campylobacter jejuni/isolamento & purificação , Campylobacter jejuni/metabolismo , Ciclo Celular , Linhagem Celular , Movimento Celular , Galinhas/microbiologia , Clatrina/farmacologia , Endocitose , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Regulação Bacteriana da Expressão Gênica , Humanos , Intestinos/microbiologia , Proteoma/análise , Espécies Reativas de Oxigênio , Transdução de Sinais , Suínos , Fatores de Virulência/metabolismo
9.
Artigo em Inglês | MEDLINE | ID: mdl-28725639

RESUMO

Anaplasma phagocytophilum transmembrane and surface proteins play a role during infection and multiplication in host neutrophils and tick vector cells. Recently, A. phagocytophilum Major surface protein 4 (MSP4) and Heat shock protein 70 (HSP70) were shown to be localized on the bacterial membrane, with a possible role during pathogen infection in ticks. In this study, we hypothesized that A. phagocytophilum MSP4 and HSP70 have similar functions in tick-pathogen and host-pathogen interactions. To address this hypothesis, herein we characterized the role of these bacterial proteins in interaction and infection of vertebrate host cells. The results showed that A. phagocytophilum MSP4 and HSP70 are involved in host-pathogen interactions, with a role for HSP70 during pathogen infection. The analysis of the potential protective capacity of MSP4 and MSP4-HSP70 antigens in immunized sheep showed that MSP4-HSP70 was only partially protective against pathogen infection. This limited protection may be associated with several factors, including the recognition of non-protective epitopes by IgG in immunized lambs. Nevertheless, these antigens may be combined with other candidate protective antigens for the development of vaccines for the control of human and animal granulocytic anaplasmosis. Focusing on the characterization of host protective immune mechanisms and protein-protein interactions at the host-pathogen interface may lead to the discovery and design of new effective protective antigens.


Assuntos
Anaplasma phagocytophilum/metabolismo , Proteínas de Bactérias/metabolismo , Ehrlichiose/veterinária , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Membrana/metabolismo , Doenças dos Ovinos/microbiologia , Anaplasma phagocytophilum/genética , Animais , Proteínas de Bactérias/genética , Ehrlichiose/microbiologia , Proteínas de Choque Térmico HSP70/genética , Interações Hospedeiro-Patógeno , Proteínas de Membrana/genética , Ovinos
10.
Artigo em Inglês | MEDLINE | ID: mdl-28439499

RESUMO

Ticks and the pathogens they transmit constitute a growing burden for human and animal health worldwide. Vector competence is a component of vectorial capacity and depends on genetic determinants affecting the ability of a vector to transmit a pathogen. These determinants affect traits such as tick-host-pathogen and susceptibility to pathogen infection. Therefore, the elucidation of the mechanisms involved in tick-pathogen interactions that affect vector competence is essential for the identification of molecular drivers for tick-borne diseases. In this review, we provide a comprehensive overview of tick-pathogen molecular interactions for bacteria, viruses, and protozoa affecting human and animal health. Additionally, the impact of tick microbiome on these interactions was considered. Results show that different pathogens evolved similar strategies such as manipulation of the immune response to infect vectors and facilitate multiplication and transmission. Furthermore, some of these strategies may be used by pathogens to infect both tick and mammalian hosts. Identification of interactions that promote tick survival, spread, and pathogen transmission provides the opportunity to disrupt these interactions and lead to a reduction in tick burden and the prevalence of tick-borne diseases. Targeting some of the similar mechanisms used by the pathogens for infection and transmission by ticks may assist in development of preventative strategies against multiple tick-borne diseases.


Assuntos
Vetores Aracnídeos/microbiologia , Vetores Aracnídeos/virologia , Transmissão de Doença Infecciosa , Interações Hospedeiro-Patógeno , Doenças Transmitidas por Carrapatos/epidemiologia , Carrapatos/fisiologia , Animais , Vetores Aracnídeos/parasitologia , Humanos , Carrapatos/microbiologia , Carrapatos/parasitologia , Carrapatos/virologia
11.
Epigenetics ; 11(4): 303-19, 2016 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-27019326

RESUMO

Epigenetic mechanisms have not been characterized in ticks despite their importance as vectors of human and animal diseases worldwide. The objective of this study was to characterize the histones and histone modifying enzymes (HMEs) of the tick vector Ixodes scapularis and their role during Anaplasma phagocytophilum infection. We first identified 5 histones and 34 HMEs in I. scapularis in comparison with similar proteins in model organisms. Then, we used transcriptomic and proteomic data to analyze the mRNA and protein levels of I. scapularis histones and HMEs in response to A. phagocytophilum infection of tick tissues and cultured cells. Finally, selected HMEs were functionally characterized by pharmacological studies in cultured tick cells. The results suggest that A. phagocytophilum manipulates tick cell epigenetics to increase I. scapularis p300/CBP, histone deacetylase, and Sirtuin levels, resulting in an inhibition of cell apoptosis that in turn facilitates pathogen infection and multiplication. These results also suggest that a compensatory mechanism might exist by which A. phagocytophilum manipulates tick HMEs to regulate transcription and apoptosis in a tissue-specific manner to facilitate infection, but preserving tick fitness to guarantee survival of both pathogens and ticks. Our study also indicates that the pathogen manipulates arthropod and vertebrate cell epigenetics in similar ways to inhibit the host response to infection. Epigenetic regulation of tick biological processes is an essential element of the infection by A. phagocytophilum and the study of the mechanisms and principal actors involved is likely to provide clues for the development of anti-tick drugs and vaccines.


Assuntos
Anaplasma phagocytophilum/genética , Epigênese Genética , Código das Histonas/genética , Histonas/genética , Animais , Apoptose/genética , Linhagem Celular , Histonas/metabolismo , Interações Hospedeiro-Patógeno/genética , Humanos , Insetos Vetores/genética , Ixodes/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Transcriptoma/genética , Fatores de Transcrição de p300-CBP/biossíntese , Fatores de Transcrição de p300-CBP/genética
12.
Artigo em Inglês | MEDLINE | ID: mdl-26904518

RESUMO

Anaplasma phagocytophilum are transmitted by Ixodes spp. ticks and have become one of the most common and relevant tick-borne pathogens due to their impact on human and animal health. Recent results have increased our understanding of the molecular interactions between Ixodes scapularis and A. phagocytophilum through the demonstration of tissue-specific molecular pathways that ensure pathogen infection, development and transmission by ticks. However, little is known about the Ixodes ricinus genes and proteins involved in the response to A. phagocytophilum infection. The tick species I. scapularis and I. ricinus are evolutionarily closely related and therefore similar responses are expected in A. phagocytophilum-infected cells. However, differences may exist between I. scapularis ISE6 and I. ricinus IRE/CTVM20 tick cells associated with tissue-specific signatures of these cell lines. To address this hypothesis, the transcriptional response to A. phagocytophilum infection was characterized by RNA sequencing and compared between I. scapularis ISE6 and I. ricinus IRE/CTVM20 tick cell lines. The transcriptional response to infection of I. scapularis ISE6 cells resembled that of tick hemocytes while the response in I. ricinus IRE/CTVM20 cells was more closely related to that reported previously in infected tick midguts. The inhibition of cell apoptosis by A. phagocytophilum appears to be a key adaptation mechanism to facilitate infection of both vertebrate and tick cells and was used to investigate further the tissue-specific response of tick cell lines to pathogen infection. The results supported a role for the intrinsic pathway in the inhibition of cell apoptosis by A. phagocytophilum infection of I. scapularis ISE6 cells. In contrast, the results in I. ricinus IRE/CTVM20 cells were similar to those obtained in tick midguts and suggested a role for the JAK/STAT pathway in the inhibition of apoptosis in tick cells infected with A. phagocytophilum. Nevertheless, tick cell lines were derived from embryonated eggs and may contain various cell populations with different morphology and behavior that could affect transcriptional response to infection. These results suggested tissue-specific signatures in I. scapularis ISE6 and I. ricinus IRE/CTVM20 tick cell line response to A. phagocytophilum infection that support their use as models for the study of tick-pathogen interactions.


Assuntos
Anaplasma phagocytophilum/patogenicidade , Apoptose/genética , Interações Hospedeiro-Patógeno/genética , Ixodes/microbiologia , RNA/genética , Animais , Sequência de Bases , Linhagem Celular , Regulação da Expressão Gênica/genética , Análise de Sequência de RNA , Transcrição Gênica/genética
13.
Parasit Vectors ; 9: 467, 2016 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-27561965

RESUMO

BACKGROUND: The intracellular bacteria Anaplasma phagocytophilum are emerging zoonotic pathogens affecting human and animal health, and a good model for the study of tick-host-pathogen interactions. This tick-borne pathogen is transmitted by Ixodes scapularis in the United States where it causes human granulocytic anaplasmosis. Tick midguts and salivary glands play a major role during tick feeding and development, and in pathogen acquisition, multiplication and transmission. Vertebrate host proteins are found in tick midguts after feeding and have been described in the salivary glands of fed and unfed ticks, suggesting a role for these proteins during tick feeding and development. Furthermore, recent results suggested the hypothesis that pathogen infection affects tick metabolic processes to modify host protein digestion and persistence in the tick with possible implications for tick physiology and pathogen life-cycle. METHODS: To address this hypothesis, herein we used I. scapularis female ticks fed on uninfected and A. phagocytophilum-infected sheep to characterize host protein content in midguts and salivary glands by proteomic analysis of tick tissues. RESULTS: The results evidenced a clear difference in the host protein content between tick midguts and salivary glands in response to infection suggesting that A. phagocytophilum selectively manipulates the levels of vertebrate host proteins in ticks in a tissue-specific manner to facilitate pathogen infection, multiplication and transmission while preserving tick feeding and development. The mechanisms by which A. phagocytophilum manipulates the levels of vertebrate host proteins are not known, but the results obtained here suggested that it might include the modification of proteolytic pathways. CONCLUSIONS: The results of this study provided evidence to support that A. phagocytophilum affect tick proteolytic pathways to selectively manipulate the levels of vertebrate host proteins in a tissue-specific manner to increase tick vector capacity. Investigating the biological relevance of host proteins in tick biology and pathogen infection and the mechanisms used by A. phagocytophilum to manipulate host protein content is essential to advance our knowledge of tick-host-pathogen molecular interactions. These results have implications for the identification of new targets for the development of vaccines for the control of tick-borne diseases.


Assuntos
Anaplasma phagocytophilum/fisiologia , Ixodes/metabolismo , Ixodes/microbiologia , Ovinos/metabolismo , Animais , Proteínas de Artrópodes/genética , Proteínas de Artrópodes/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Interações Hospedeiro-Patógeno , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
14.
Nat Commun ; 7: 10507, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26856261

RESUMO

Ticks transmit more pathogens to humans and animals than any other arthropod. We describe the 2.1 Gbp nuclear genome of the tick, Ixodes scapularis (Say), which vectors pathogens that cause Lyme disease, human granulocytic anaplasmosis, babesiosis and other diseases. The large genome reflects accumulation of repetitive DNA, new lineages of retro-transposons, and gene architecture patterns resembling ancient metazoans rather than pancrustaceans. Annotation of scaffolds representing ∼57% of the genome, reveals 20,486 protein-coding genes and expansions of gene families associated with tick-host interactions. We report insights from genome analyses into parasitic processes unique to ticks, including host 'questing', prolonged feeding, cuticle synthesis, blood meal concentration, novel methods of haemoglobin digestion, haem detoxification, vitellogenesis and prolonged off-host survival. We identify proteins associated with the agent of human granulocytic anaplasmosis, an emerging disease, and the encephalitis-causing Langat virus, and a population structure correlated to life-history traits and transmission of the Lyme disease agent.


Assuntos
Anaplasma phagocytophilum , Vetores Aracnídeos/genética , Genoma/genética , Ixodes/genética , Canais Iônicos de Abertura Ativada por Ligante/genética , Animais , Perfilação da Expressão Gênica , Genômica , Doença de Lyme/transmissão , Oócitos , Xenopus laevis
15.
Ticks Tick Borne Dis ; 6(6): 758-67, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26183310

RESUMO

Anaplasma phagocytophilum is an intracellular rickettsial pathogen transmitted by Ixodes spp. ticks, which causes granulocytic anaplasmosis in humans, horses and dogs and tick-borne fever (TBF) in ruminants. In the United States, human granulocytic anaplasmosis (HGA) is highly prevalent while TBF has not been reported. However, in Europe the situation is the opposite, with high prevalence for TBF in sheep and low prevalence of HGA. The origin of these differences has not been identified and our hypothesis is that different A. phagocytophilum isolates impact differently on tick vector capacity through inhibition of apoptosis to establish infection of the tick vector. In this study we used three different isolates of A. phagocytophilum of human, canine and ovine origin to infect the Ixodes ricinus-derived cell line IRE/CTVM20 and the Ixodes scapularis-derived cell line ISE6 in order to characterize the effect of infection on the level of tick cell apoptosis. Inhibition of apoptosis was observed by flow cytometry as early as 24h post-infection for both tick cell lines and all three isolates of A. phagocytophilum, suggesting that pathogen infection inhibits apoptotic pathways to facilitate infection independently of the origin of the A. phagocytophilum isolate and tick vector species. However, infection with A. phagocytophilum isolates inhibited the intrinsic apoptosis pathway at different levels in I. scapularis and I. ricinus cells. These results suggested an impact of vector-pathogen co-evolution on the adaptation of A. phagocytophilum isolates to grow in tick cells as each isolate grew better in the tick cell line derived from its natural vector species. These results increase our understanding of the mechanisms of A. phagocytophilum infection and multiplication and suggest that multiple mechanisms may affect disease prevalence in different geographical regions.


Assuntos
Anaplasma phagocytophilum/fisiologia , Apoptose/fisiologia , Ixodes/citologia , Animais , Caspase 9/genética , Caspase 9/metabolismo , Linhagem Celular , DNA Bacteriano/genética , Regulação Enzimológica da Expressão Gênica , Hexoquinase/genética , Hexoquinase/metabolismo , Ixodes/embriologia , Filogenia
16.
PLoS One ; 10(7): e0133038, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26186700

RESUMO

Tudor staphylococcal nuclease (Tudor-SN) and Argonaute (Ago) are conserved components of the basic RNA interference (RNAi) machinery with a variety of functions including immune response and gene regulation. The RNAi machinery has been characterized in tick vectors of human and animal diseases but information is not available on the role of Tudor-SN in tick RNAi and other cellular processes. Our hypothesis is that tick Tudor-SN is part of the RNAi machinery and may be involved in innate immune response and other cellular processes. To address this hypothesis, Ixodes scapularis and I. ricinus ticks and/or cell lines were used to annotate and characterize the role of Tudor-SN in dsRNA-mediated RNAi, immune response to infection with the rickettsia Anaplasma phagocytophilum and the flaviviruses TBEV or LGTV and tick feeding. The results showed that Tudor-SN is conserved in ticks and involved in dsRNA-mediated RNAi and tick feeding but not in defense against infection with the examined viral and rickettsial pathogens. The effect of Tudor-SN gene knockdown on tick feeding could be due to down-regulation of genes that are required for protein processing and blood digestion through a mechanism that may involve selective degradation of dsRNAs enriched in G:U pairs that form as a result of adenosine-to-inosine RNA editing. These results demonstrated that Tudor-SN plays a role in tick RNAi pathway and feeding but no strong evidence for a role in innate immune responses to pathogen infection was found.


Assuntos
Anaplasma phagocytophilum/patogenicidade , Flavivirus/patogenicidade , Ixodes/genética , Proteínas Nucleares/genética , Interferência de RNA , Sequência de Aminoácidos , Animais , Linhagem Celular , Sequência Conservada , Cricetinae , Ixodes/parasitologia , Ixodes/virologia , Dados de Sequência Molecular , Proteínas Nucleares/metabolismo , Filogenia , Transcriptoma
17.
Ticks Tick Borne Dis ; 6(6): 820-8, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26219233

RESUMO

Ticks are vectors of diseases that affect humans and animals worldwide. Tick vaccines have been proposed as a cost-effective and environmentally sound alternative for tick control. Recently, the Rhipicephalus microplus Subolesin (SUB)-Anaplasma marginale MSP1a chimeric antigen was produced in Escherichia coli as membrane-bound and exposed protein and used to protect vaccinated cattle against tick infestations. In this research, lipidomics and proteomics characterization of the E. coli membrane-bound SUB-MSP1a antigen showed the presence of components with potential adjuvant effect. Furthermore, vaccination with membrane-free SUB-MSP1a and bacterial membranes containing SUB-MSP1a showed that bacterial membranes enhance the immunogenicity of the SUB-MSP1a antigen in animal models. R. microplus female ticks were capillary-fed with sera from pigs orally immunized with membrane-free SUB, membrane bound SUB-MSP1a and saline control. Ticks ingested antibodies added to the blood meal and the effect of these antibodies on reduction of tick weight was shown for membrane bound SUB-MSP1a but not SUB when compared to control. Using the simple and cost-effective process developed for the purification of membrane-bound SUB-MSP1a, endotoxin levels were within limits accepted for recombinant vaccines. These results provide further support for the development of tick vaccines using E. coli membranes exposing chimeric antigens such as SUB-MSP1a.


Assuntos
Anaplasma marginale/imunologia , Antígenos/imunologia , Proteínas de Artrópodes/imunologia , Proteínas da Membrana Bacteriana Externa/imunologia , Rhipicephalus/imunologia , Vacinas/imunologia , Adjuvantes Imunológicos , Animais , Anticorpos/sangue , Membrana Celular/química , Escherichia coli , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Coelhos , Suínos
18.
Ticks Tick Borne Dis ; 6(4): 522-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25943785

RESUMO

Anaplasma marginale is an economically important tick-borne pathogen of cattle that causes bovine anaplasmosis. A wide range of geographic strains of A. marginale have been isolated from cattle, several of which have been characterized using genomics and proteomics. While many of these strains have been propagated in tick lines, comparative analyses after propagation in tick cells have not been reported. The overall purpose of this research therefore was to compare the degree of conservation of selected genes after propagation in tick cell culture among A. marginale strains from the U.S. (the Virginia strain) and Brazil (UFMG1 and UFMG2 strains). The genes studied herein included those which encode the proteins HSP70 and SODB involved in heat shock and stress responses, respectively, and two genes that encode major surface proteins MSP4 and MSP5. Strain identities were first confirmed by sequencing the tandem repeats of the msp1a gene which encodes for the adhesin, MSP1a. The results of these studies demonstrated that the genes encoding for both stress response and heat shock proteins were highly conserved among the three A. marginale strains. Antibodies specific for MSP4, MSP5, SODB and HSP70 proteins were used to further characterize the A. marginale strains, and they reacted with all of these strains propagated in tick cell culture, providing further evidence for antigenic conservation. Although antigenic differences were not found among the three A. marginale strains, multi-locus sequence analysis (MLSA) performed with nucleotide sequences of these genes demonstrated that the A. marginale Brazilian and U.S. strains fall in different clades. These results showed that phylogenetically distant strains of A. marginale are antigenically conserved, even after several in vitro passages, supporting the use of some of the above conserved proteins as candidates for universal vaccines.


Assuntos
Anaplasma marginale/isolamento & purificação , Anaplasmose/imunologia , Vetores Aracnídeos/microbiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Doenças dos Bovinos/imunologia , Carrapatos/microbiologia , Anaplasma marginale/classificação , Anaplasma marginale/genética , Anaplasma marginale/crescimento & desenvolvimento , Anaplasmose/microbiologia , Animais , Variação Antigênica , Brasil , Bovinos , Doenças dos Bovinos/microbiologia , Sequência Conservada , Dados de Sequência Molecular , Filogenia , Estados Unidos
19.
PLoS One ; 10(9): e0137237, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26340562

RESUMO

Anaplasma phagocytophilum is an emerging zoonotic pathogen transmitted by Ixodes scapularis that causes human granulocytic anaplasmosis. Here, a high throughput quantitative proteomics approach was used to characterize A. phagocytophilum proteome during rickettsial multiplication and identify proteins involved in infection of the tick vector, I. scapularis. The first step in this research was focused on tick cells infected with A. phagocytophilum and sampled at two time points containing 10-15% and 65-71% infected cells, respectively to identify key bacterial proteins over-represented in high percentage infected cells. The second step was focused on adult female tick guts and salivary glands infected with A. phagocytophilum to compare in vitro results with those occurring during bacterial infection in vivo. The results showed differences in the proteome of A. phagocytophilum in infected ticks with higher impact on protein synthesis and processing than on bacterial replication in tick salivary glands. These results correlated well with the developmental cycle of A. phagocytophilum, in which cells convert from an intracellular reticulated, replicative form to the nondividing infectious dense-core form. The analysis of A. phagocytophilum differentially represented proteins identified stress response (GroEL, HSP70) and surface (MSP4) proteins that were over-represented in high percentage infected tick cells and salivary glands when compared to low percentage infected cells and guts, respectively. The results demonstrated that MSP4, GroEL and HSP70 interact and bind to tick cells, thus playing a role in rickettsia-tick interactions. The most important finding of these studies is the increase in the level of certain bacterial stress response and surface proteins in A. phagocytophilum-infected tick cells and salivary glands with functional implication in tick-pathogen interactions. These results gave a new dimension to the role of these stress response and surface proteins during A. phagocytophilum infection in ticks. Characterization of Anaplasma proteome contributes information on host-pathogen interactions and provides targets for development of novel control strategies for pathogen infection and transmission.


Assuntos
Proteínas de Bactérias/genética , Chaperonina 60/genética , Proteínas de Choque Térmico HSP70/genética , Ixodes/microbiologia , Proteínas de Membrana/genética , Proteoma/genética , Anaplasma phagocytophilum , Animais , Proteínas de Bactérias/metabolismo , Chaperonina 60/metabolismo , Feminino , Trato Gastrointestinal/microbiologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Proteínas de Choque Térmico HSP70/metabolismo , Interações Hospedeiro-Patógeno , Proteínas de Membrana/metabolismo , Anotação de Sequência Molecular , Proteoma/metabolismo , Glândulas Salivares/microbiologia , Transdução de Sinais , Estresse Fisiológico
20.
PLoS One ; 9(2): e89564, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24586875

RESUMO

BACKGROUND: Dermacentor reticulatus (Fabricius, 1794) is distributed in Europe and Asia where it infests and transmits disease-causing pathogens to humans, pets and other domestic and wild animals. However, despite its role as a vector of emerging or re-emerging diseases, very little information is available on the genome, transcriptome and proteome of D. reticulatus. Tick larvae are the first developmental stage to infest hosts, acquire infection and transmit pathogens that are transovarially transmitted and are exposed to extremely stressing conditions. In this study, we used a systems biology approach to get an insight into the mechanisms active in D. reticulatus unfed larvae, with special emphasis on stress response. PRINCIPAL FINDINGS: The results support the use of paired end RNA sequencing and proteomics informed by transcriptomics (PIT) for the analysis of transcriptomics and proteomics data, particularly for organisms such as D. reticulatus with little sequence information available. The results showed that metabolic and cellular processes involved in protein synthesis were the most active in D. reticulatus unfed larvae, suggesting that ticks are very active during this life stage. The stress response was activated in D. reticulatus unfed larvae and a Rickettsia sp. similar to R. raoultii was identified in these ticks. SIGNIFICANCE: The activation of stress responses in D. reticulatus unfed larvae likely counteracts the negative effect of temperature and other stress conditions such as Rickettsia infection and favors tick adaptation to environmental conditions to increase tick survival. These results show mechanisms that have evolved in D. reticulatus ticks to survive under stress conditions and suggest that these mechanisms are conserved across hard tick species. Targeting some of these proteins by vaccination may increase tick susceptibility to natural stress conditions, which in turn reduce tick survival and reproduction, thus reducing tick populations and vector capacity for tick-borne pathogens.


Assuntos
Vetores Aracnídeos/fisiologia , Dermacentor/fisiologia , Estresse Fisiológico , Animais , Vetores Aracnídeos/microbiologia , Proteínas de Artrópodes/genética , Proteínas de Artrópodes/metabolismo , Dermacentor/microbiologia , Privação de Alimentos , Genes Bacterianos , Larva/microbiologia , Larva/fisiologia , Biossíntese de Proteínas , Proteoma/genética , Proteoma/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Rickettsia/genética , Biologia de Sistemas , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA