Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 12: 636623, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34025644

RESUMO

The inflammatory response to viral infection in humans is a dynamic process with complex cell interactions that are governed by the immune system and influenced by both host and viral factors. Due to this complexity, the relative contributions of the virus and host factors are best studied in vivo using animal models. In this review, we describe how the zebrafish (Danio rerio) has been used as a powerful model to study host-virus interactions and inflammation by combining robust forward and reverse genetic tools with in vivo imaging of transparent embryos and larvae. The innate immune system has an essential role in the initial inflammatory response to viral infection. Focused studies of the innate immune response to viral infection are possible using the zebrafish model as there is a 4-6 week timeframe during development where they have a functional innate immune system dominated by neutrophils and macrophages. During this timeframe, zebrafish lack a functional adaptive immune system, so it is possible to study the innate immune response in isolation. Sequencing of the zebrafish genome has revealed significant genetic conservation with the human genome, and multiple studies have revealed both functional conservation of genes, including those critical to host cell infection and host cell inflammatory response. In addition to studying several fish viruses, zebrafish infection models have been developed for several human viruses, including influenza A, noroviruses, chikungunya, Zika, dengue, herpes simplex virus type 1, Sindbis, and hepatitis C virus. The development of these diverse viral infection models, coupled with the inherent strengths of the zebrafish model, particularly as it relates to our understanding of macrophage and neutrophil biology, offers opportunities for far more intensive studies aimed at understanding conserved host responses to viral infection. In this context, we review aspects relating to the evolution of innate immunity, including the evolution of viral pattern recognition receptors, interferons and interferon receptors, and non-coding RNAs.


Assuntos
Inflamação/imunologia , Viroses/imunologia , Peixe-Zebra/imunologia , Animais , Homeostase , Imunidade Inata , Controle de Infecções
2.
PLoS One ; 12(6): e0179517, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28658311

RESUMO

Comparative functional genomic studies require the proper identification of gene orthologs to properly exploit animal biomedical research models. To identify gene orthologs, comprehensive, conserved gene synteny analyses are necessary to unwind gene histories that are convoluted by two rounds of early vertebrate genome duplication, and in the case of the teleosts, a third round, the teleost genome duplication (TGD). Recently, the genome of the spotted gar, a holostean outgroup to the teleosts that did not undergo this third genome duplication, was sequenced and applied as an orthology bridge to facilitate the identification of teleost orthologs to human genes and to enhance the power of teleosts as biomedical models. In this study, we apply the spotted gar orthology bridge to help describe the gene history of the vertebrate TNFAIP8 family. Members of the TNFAIP8 gene family have been linked to regulation of immune function and homeostasis and the development of multiple cancer types. Through a conserved gene synteny analysis, we identified zebrafish orthologs to human TNFAIP8L1 and TNFAIP8L3 genes and two co-orthologs to human TNFAIP8L2, but failed to identify an ortholog to human TNFAIP8. Through the application of the orthology bridge, we determined that teleost orthologs to human TNFAIP8 genes were likely lost in a genome inversion event after their divergence from their common ancestor with spotted gar. These findings demonstrate the value of this enhanced approach to gene history analysis and support the development of teleost models to study complex questions related to an array of biomedical issues, including immunity and cancer.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Evolução Molecular , Peixes/genética , Sintenia , Animais , Bases de Dados Genéticas , Humanos , Filogenia
3.
J Vis Exp ; (119)2017 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-28190053

RESUMO

Each year, seasonal influenza outbreaks profoundly affect societies worldwide. In spite of global efforts, influenza remains an intractable healthcare burden. The principle strategy to curtail infections is yearly vaccination. In individuals who have contracted influenza, antiviral drugs can mitigate symptoms. There is a clear and unmet need to develop alternative strategies to combat influenza. Several animal models have been created to model host-influenza interactions. Here, protocols for generating zebrafish models for systemic and localized human influenza A virus (IAV) infection are described. Using a systemic IAV infection model, small molecules with potential antiviral activity can be screened. As a proof-of-principle, a protocol that demonstrates the efficacy of the antiviral drug Zanamivir in IAV-infected zebrafish is described. It shows how disease phenotypes can be quantified to score the relative efficacy of potential antivirals in IAV-infected zebrafish. In recent years, there has been increased appreciation for the critical role neutrophils play in the human host response to influenza infection. The zebrafish has proven to be an indispensable model for the study of neutrophil biology, with direct impacts on human medicine. A protocol to generate a localized IAV infection in the Tg(mpx:mCherry) zebrafish line to study neutrophil biology in the context of a localized viral infection is described. Neutrophil recruitment to localized infection sites provides an additional quantifiable phenotype for assessing experimental manipulations that may have therapeutic applications. Both zebrafish protocols described faithfully recapitulate aspects of human IAV infection. The zebrafish model possesses numerous inherent advantages, including high fecundity, optical clarity, amenability to drug screening, and availability of transgenic lines, including those in which immune cells such as neutrophils are labeled with fluorescent proteins. The protocols detailed here exploit these advantages and have the potential to reveal critical insights into host-IAV interactions that may ultimately translate into the clinic.


Assuntos
Antivirais/farmacologia , Neutrófilos/imunologia , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções por Orthomyxoviridae/imunologia , Animais , Modelos Animais de Doenças , Humanos , Vírus da Influenza A , Infecções por Orthomyxoviridae/veterinária , Zanamivir/farmacologia , Peixe-Zebra
4.
J Appl Toxicol ; 36(12): 1662-1667, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27111768

RESUMO

Triclosan (TCS) is a synthetic antimicrobial agent used in many consumer goods at millimolar concentrations. As a result of exposure, TCS has been detected widely in humans. We have recently discovered that TCS is a proton ionophore mitochondrial uncoupler in multiple types of living cells. Here, we present novel data indicating that TCS is also a mitochondrial uncoupler in a living organism: 24-hour post-fertilization (hpf) zebrafish embryos. These experiments were conducted using a Seahorse Bioscience XFe 96 Extracellular Flux Analyzer modified for bidirectional temperature control, using the XF96 spheroid plate to position and measure one zebrafish embryo per well. Using this method, after acute exposure to TCS, the basal oxygen consumption rate (OCR) increases, without a decrease in survival or heartbeat rate. TCS also decreases ATP-linked respiration and spare respiratory capacity and increases proton leak: all indicators of mitochondrial uncoupling. Our data indicate, that TCS is a mitochondrial uncoupler in vivo, which should be taken into consideration when assessing the toxicity and/or pharmaceutical uses of TCS. This is the first example of usage of a Seahorse Extracellular Flux Analyzer to measure bioenergetic flux of a single zebrafish embryo per well in a 96-well assay format. The method developed in this study provides a high-throughput tool to identify previously unknown mitochondrial uncouplers in a living organism. Copyright © 2016 John Wiley & Sons, Ltd.


Assuntos
Embrião não Mamífero/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Mitocôndrias/efeitos dos fármacos , Triclosan/toxicidade , Desacopladores/toxicidade , Peixe-Zebra , Animais , Relação Dose-Resposta a Droga , Mitocôndrias/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Prótons , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo
5.
PLoS One ; 10(2): e0117225, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25646724

RESUMO

Light microscopy enables noninvasive imaging of fluorescent species in biological specimens, but resolution is generally limited by diffraction to ~200-250 nm. Many biological processes occur on smaller length scales, highlighting the importance of techniques that can image below the diffraction limit and provide valuable single-molecule information. In recent years, imaging techniques have been developed which can achieve resolution below the diffraction limit. Utilizing one such technique, fluorescence photoactivation localization microscopy (FPALM), we demonstrated its ability to construct super-resolution images from single molecules in a living zebrafish embryo, expanding the realm of previous super-resolution imaging to a living vertebrate organism. We imaged caveolin-1 in vivo, in living zebrafish embryos. Our results demonstrate the successful image acquisition of super-resolution images in a living vertebrate organism, opening several opportunities to answer more dynamic biological questions in vivo at the previously inaccessible nanoscale.


Assuntos
Caveolina 1/química , Membrana Celular/metabolismo , Microscopia de Fluorescência/métodos , Nanotecnologia/métodos , Animais , Caveolina 1/metabolismo , Estrutura Terciária de Proteína , Transporte Proteico , Peixe-Zebra
6.
Dis Model Mech ; 7(11): 1227-37, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25190709

RESUMO

Seasonal influenza virus infections cause annual epidemics and sporadic pandemics. These present a global health concern, resulting in substantial morbidity, mortality and economic burdens. Prevention and treatment of influenza illness is difficult due to the high mutation rate of the virus, the emergence of new virus strains and increasing antiviral resistance. Animal models of influenza infection are crucial to our gaining a better understanding of the pathogenesis of and host response to influenza infection, and for screening antiviral compounds. However, the current animal models used for influenza research are not amenable to visualization of host-pathogen interactions or high-throughput drug screening. The zebrafish is widely recognized as a valuable model system for infectious disease research and therapeutic drug testing. Here, we describe a zebrafish model for human influenza A virus (IAV) infection and show that zebrafish embryos are susceptible to challenge with both influenza A strains APR8 and X-31 (Aichi). Influenza-infected zebrafish show an increase in viral burden and mortality over time. The expression of innate antiviral genes, the gross pathology and the histopathology in infected zebrafish recapitulate clinical symptoms of influenza infections in humans. This is the first time that zebrafish embryos have been infected with a fluorescent IAV in order to visualize infection in a live vertebrate host, revealing a pattern of vascular endothelial infection. Treatment of infected zebrafish with a known anti-influenza compound, Zanamivir, reduced mortality and the expression of a fluorescent viral gene product, demonstrating the validity of this model to screen for potential antiviral drugs. The zebrafish model system has provided invaluable insights into host-pathogen interactions for a range of infectious diseases. Here, we demonstrate a novel use of this species for IAV research. This model has great potential to advance our understanding of influenza infection and the associated host innate immune response.


Assuntos
Antivirais/uso terapêutico , Modelos Animais de Doenças , Vírus da Influenza A/isolamento & purificação , Influenza Humana/virologia , Animais , Humanos , Vírus da Influenza A/fisiologia , Influenza Humana/tratamento farmacológico , Replicação Viral , Peixe-Zebra/embriologia
7.
Dev Comp Immunol ; 46(1): 84-95, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24718256

RESUMO

Humans and viruses have a long co-evolutionary history. Viral illnesses have and will continue to shape human history: from smallpox, to influenza, to HIV, and beyond. Animal models of human viral illnesses are needed in order to generate safe and effective antiviral medicines, adjuvant therapies, and vaccines. These animal models must support the replication of human viruses, recapitulate aspects of human viral illnesses, and respond with conserved immune signaling cascades. The zebrafish is perhaps the simplest, most commonly used laboratory model organism in which innate and/or adaptive immunity can be studied. Herein, we will discuss the current zebrafish models of human viral illnesses and the insights they have provided. We will highlight advantages of early life stage zebrafish and the importance of innate immunity in human viral illnesses. We will also discuss viral characteristics to consider before infecting zebrafish with human viruses as well as predict other human viruses that may be able to infect zebrafish.


Assuntos
Modelos Animais de Doenças , Viroses/imunologia , Peixe-Zebra/imunologia , Animais , Humanos , Interferons/imunologia , Viroses/virologia , Peixe-Zebra/crescimento & desenvolvimento
8.
Immunogenetics ; 66(4): 267-79, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24469064

RESUMO

The polymeric immunoglobulin (Ig) receptor (pIgR) is an integral transmembrane glycoprotein that plays an important role in the mammalian immune response by transporting soluble polymeric Igs across mucosal epithelial cells. Single pIgR genes, which are expressed in lymphoid organs including mucosal tissues, have been identified in several teleost species. A single pigr gene has been identified on zebrafish chromosome 2 along with a large multigene family consisting of 29 pigr-like (PIGRL) genes. Full-length transcripts from ten different PIGRL genes that encode secreted and putative inhibitory membrane-bound receptors have been characterized. Although PIGRL and pigr transcripts are detected in immune tissues, only PIGRL transcripts can be detected in lymphoid and myeloid cells. In contrast to pIgR which binds Igs, certain PIGRL proteins bind phospholipids. PIGRL transcript levels are increased after infection with Streptococcus iniae, suggesting a role for PIGRL genes during bacterial challenge. Transcript levels of PIGRL genes are decreased after infection with Snakehead rhabdovirus, suggesting that viral infection may suppress PIGRL function.


Assuntos
Receptores de Imunoglobulina Polimérica/genética , Receptores de Imunoglobulina Polimérica/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/imunologia , Peixe-Zebra/genética , Peixe-Zebra/imunologia , Sequência de Aminoácidos , Animais , Mapeamento Cromossômico , Sequência Conservada , Evolução Molecular , Peixes/genética , Peixes/imunologia , Expressão Gênica , Humanos , Imunidade Inata/genética , Ligantes , Mamíferos/genética , Mamíferos/imunologia , Dados de Sequência Molecular , Família Multigênica , Fosfolipídeos/metabolismo , Filogenia , Ligação Proteica , Estrutura Terciária de Proteína , Receptores de Imunoglobulina Polimérica/química , Infecções por Rhabdoviridae/genética , Infecções por Rhabdoviridae/imunologia , Infecções por Rhabdoviridae/metabolismo , Homologia de Sequência de Aminoácidos , Infecções Estreptocócicas/genética , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/metabolismo , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
9.
J Vis Exp ; (79)2013 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-24056405

RESUMO

The phagocyte respiratory burst is part of the innate immune response to pathogen infection and involves the production of reactive oxygen species (ROS). ROS are toxic and function to kill phagocytized microorganisms. In vivo quantification of phagocyte-derived ROS provides information regarding an organism's ability to mount a robust innate immune response. Here we describe a protocol to quantify and compare ROS in whole zebrafish embryos upon chemical induction of the phagocyte respiratory burst. This method makes use of a non-fluorescent compound that becomes fluorescent upon oxidation by ROS. Individual zebrafish embryos are pipetted into the wells of a microplate and incubated in this fluorogenic substrate with or without a chemical inducer of the respiratory burst. Fluorescence in each well is quantified at desired time points using a microplate reader. Fluorescence readings are adjusted to eliminate background fluorescence and then compared using an unpaired t-test. This method allows for comparison of the respiratory burst potential of zebrafish embryos at different developmental stages and in response to experimental manipulations such as protein knockdown, overexpression, or treatment with pharmacological agents. This method can also be used to monitor the respiratory burst response in whole dissected kidneys or cell preparations from kidneys of adult zebrafish and some other fish species. We believe that the relative simplicity and adaptability of this protocol will complement existing protocols and will be of interest to researchers who seek to better understand the innate immune response.


Assuntos
Medições Luminescentes/métodos , Explosão Respiratória/imunologia , Peixe-Zebra/imunologia , Animais , Embrião não Mamífero , Fluoresceínas/química , Fluoresceínas/metabolismo , Fluorescência , Imunidade Inata/imunologia , Oxirredução , Espécies Reativas de Oxigênio/imunologia , Espécies Reativas de Oxigênio/metabolismo
10.
PLoS One ; 8(7): e68759, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23874753

RESUMO

Understanding spatial distribution and dynamics of receptors within unperturbed membranes is essential for elucidating their role in antiviral signaling, but conventional studies of detergent-resistant membrane fractions cannot provide this information. Caveolae are integral to numerous signaling pathways and these membrane domains have been previously implicated in viral entry but not antiviral defense. This study shows, for the first time, the importance of spatio-temporal regulation of signaling receptors and the importance of the regulation of clustering for downstream signaling. A novel mechanism for virus evasion of host cell defenses is demonstrated through disruption of clusters of signaling molecules organized within caveolin-rich domains. Viral infection leads to a downregulation in Caveolin-1b (Cav-1b), disrupting clusters of CRFB1, a zebrafish type I interferon receptor (-R) subunit. Super-resolution microscopy has enabled the first single-molecule imaging of CRFB1 association with cav-1b-containing membrane domains. Strikingly, downregulation of Cav-1b, the major protein component of caveolae, caused CRFB1 clusters to disperse. Dispersal of CRFB1 clusters led to a suppressed antiviral immune response both in vitro and in vivo, through abrogation of downstream signaling. This response strongly suggests that CRFB1 organization within cav-1b-containing membrane domains is critical for IFN-mediated antiviral defense and presents a previously undescribed antiviral evasion strategy to alter IFN signaling and the antiviral immune response.


Assuntos
Caveolina 1/metabolismo , Resistência à Doença , Receptores de Interferon/metabolismo , Transdução de Sinais , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Peixe-Zebra/virologia , Animais , Membrana Celular/metabolismo , Resistência à Doença/imunologia , Doenças dos Peixes/imunologia , Doenças dos Peixes/metabolismo , Doenças dos Peixes/virologia , Imunidade Inata/genética , Interferons/metabolismo , Ligação Proteica , Peixe-Zebra/imunologia
11.
Biophys J ; 104(10): 2182-92, 2013 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-23708358

RESUMO

The influenza viral membrane protein hemagglutinin (HA) is required at high concentrations on virion and host-cell membranes for infectivity. Because the role of actin in membrane organization is not completely understood, we quantified the relationship between HA and host-cell actin at the nanoscale. Results obtained using superresolution fluorescence photoactivation localization microscopy (FPALM) in nonpolarized cells show that HA clusters colocalize with actin-rich membrane regions (ARMRs). Individual molecular trajectories in live cells indicate restricted HA mobility in ARMRs, and actin disruption caused specific changes to HA clustering. Surprisingly, the actin-binding protein cofilin was excluded from some regions within several hundred nanometers of HA clusters, suggesting that HA clusters or adjacent proteins within the same clusters influence local actin structure. Thus, with the use of imaging, we demonstrate a dynamic relationship between glycoprotein membrane organization and the actin cytoskeleton at the nanoscale.


Assuntos
Actinas/metabolismo , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Citoesqueleto de Actina/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Animais , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Glicoproteínas de Hemaglutininação de Vírus da Influenza/ultraestrutura , Vírus da Influenza A Subtipo H2N2/química , Vírus da Influenza A Subtipo H2N2/metabolismo , Camundongos , Células NIH 3T3 , Multimerização Proteica
12.
Methods Cell Biol ; 105: 87-116, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21951527

RESUMO

All animals are ecosystems, home to diverse microbial populations. Animal-associated microbes play important roles in the normal development and physiology of their hosts, but can also be agents of infectious disease. Traditionally, mice have been used to study pathogenic and beneficial associations between microbes and vertebrate animals. The zebrafish is emerging as a valuable new model system for host-microbe interaction studies, affording researchers with the opportunity to survey large populations of hosts and to visualize microbe-host associations at a cellular level in living animals. This chapter provides detailed protocols for the analysis of zebrafish-associated microbial communities, the derivation and husbandry of germ-free zebrafish, and the modeling of infectious disease in different stages of zebrafish development via different routes of inoculation. These protocols offer a starting point for researchers to address a multitude of questions about animals' coexistence with microorganisms.


Assuntos
Técnicas de Tipagem Bacteriana , Biologia do Desenvolvimento/métodos , Vida Livre de Germes , Interações Hospedeiro-Patógeno , Hibridização in Situ Fluorescente/métodos , Larva , Microinjeções/métodos , Peixe-Zebra , Animais , Bactérias/crescimento & desenvolvimento , Infecções Bacterianas/embriologia , Infecções Bacterianas/microbiologia , Modelos Animais de Doenças , Embrião não Mamífero/microbiologia , Embrião não Mamífero/virologia , Intestinos/microbiologia , Intestinos/virologia , Larva/crescimento & desenvolvimento , Larva/microbiologia , Larva/virologia , Camundongos , Consórcios Microbianos , Interações Microbianas , RNA Bacteriano/análise , RNA Ribossômico 16S/análise , Viroses/embriologia , Viroses/virologia , Vírus/crescimento & desenvolvimento , Peixe-Zebra/embriologia , Peixe-Zebra/microbiologia , Peixe-Zebra/virologia
13.
Infect Immun ; 78(11): 4542-50, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20732993

RESUMO

Cystic fibrosis (CF) is a genetic disease caused by recessive mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene and is associated with prevalent and chronic Pseudomonas aeruginosa lung infections. Despite numerous studies that have sought to elucidate the role of CFTR in the innate immune response, the links between CFTR, innate immunity, and P. aeruginosa infection remain unclear. The present work highlights the zebrafish as a powerful model organism for human infectious disease, particularly infection by P. aeruginosa. Zebrafish embryos with reduced expression of the cftr gene (Cftr morphants) exhibited reduced respiratory burst response and directed neutrophil migration, supporting a connection between cftr and the innate immune response. Cftr morphants were infected with P. aeruginosa or other bacterial species that are commonly associated with infections in CF patients, including Burkholderia cenocepacia, Haemophilus influenzae, and Staphylococcus aureus. Intriguingly, the bacterial burden of P. aeruginosa was found to be significantly higher in zebrafish Cftr morphants than in controls, but this phenomenon was not observed with the other bacterial species. Bacterial burden in Cftr morphants infected with a P. aeruginosa ΔLasR mutant, a quorum sensing-deficient strain, was comparable to that in control fish, indicating that the regulation of virulence factors through LasR is required for enhancement of infection in the absence of Cftr. The zebrafish system provides a multitude of advantages for studying the pathogenesis of P. aeruginosa and for understanding the role that innate immune cells, such as neutrophils, play in the host response to acute bacterial infections commonly associated with cystic fibrosis.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Modelos Animais de Doenças , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/patogenicidade , Peixe-Zebra/imunologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Humanos , Imunidade Inata , Neutrófilos/imunologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Percepção de Quorum , Transativadores/genética , Transativadores/metabolismo , Virulência , Peixe-Zebra/microbiologia
14.
Appl Environ Microbiol ; 76(11): 3467-74, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20363780

RESUMO

To observe real-time interactions between green fluorescent protein-labeled immune cells and invading bacteria in the zebrafish (Danio rerio), a series of plasmids was constructed for the red fluorescent protein (RFP) labeling of a variety of fish and human pathogens. The aim of this study was to create a collection of plasmids that would express RFP pigments both constitutively and under tac promoter regulation and that would be nontoxic and broadly transmissible to a variety of Gram-negative bacteria. DNA fragments encoding the RFP dimeric (d), monomeric (m), and tandem dimeric (td) derivatives d-Tomato, td-Tomato, m-Orange, and m-Cherry were cloned into the IncQ-based vector pMMB66EH in Escherichia coli. Plasmids were mobilized into recipient strains by conjugal mating. Pigment production was inducible in Escherichia coli, Pseudomonas aeruginosa, Edwardsiella tarda, and Vibrio (Listonella) anguillarum strains by isopropyl-beta-d-thiogalactopyranoside (IPTG) treatment. A spontaneous mutant exconjugant of P. aeruginosa PA14 was isolated that expressed td-Tomato constitutively. Complementation analysis revealed that the constitutive phenotype likely was due to a mutation in lacI(q) carried on pMMB66EH. DNA sequence analysis confirmed the presence of five transitions, four transversions, and a 2-bp addition within a 14-bp region of lacI. Vector DNA was purified from this constitutive mutant, and structural DNA sequences for RFP pigments were cloned into the constitutive vector. Exconjugants of P. aeruginosa, E. tarda, and V. anguillarum expressed all pigments in an IPTG-independent fashion. Results from zebrafish infectivity studies indicate that RFP-labeled pathogens will be useful for the study of real-time interactions between host cells of the innate immune system and the infecting pathogen.


Assuntos
Bactérias Gram-Negativas/patogenicidade , Interações Hospedeiro-Patógeno , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Biologia Molecular/métodos , Plasmídeos , Coloração e Rotulagem/métodos , Sequência de Aminoácidos , Animais , Sequência de Bases , DNA Bacteriano/química , DNA Bacteriano/genética , Modelos Animais de Doenças , Vetores Genéticos , Bactérias Gram-Negativas/genética , Dados de Sequência Molecular , Análise de Sequência de DNA , Peixe-Zebra/microbiologia , Proteína Vermelha Fluorescente
15.
J Immunol ; 183(9): 5896-908, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19812203

RESUMO

Mammalian immune responses to LPS exposure are typified by the robust induction of NF-kappaB and IFN-beta responses largely mediated by TLR4 signal transduction pathways. In contrast to mammals, Tlr4 signal transduction pathways in nontetrapods are not well understood. Comprehensive syntenic and phylogenetic analyses support our hypothesis that zebrafish tlr4a and tlr4b genes are paralogous rather than orthologous to human TLR4. Furthermore, we provide evidence to support our assertion that the in vivo responsiveness of zebrafish to LPS exposure is not mediated by Tlr4a and Tlr4b paralogs because they fail to respond to LPS stimulation in vitro. Zebrafish Tlr4a and Tlr4b paralogs were also unresponsive to heat-killed Escherichia coli and Legionella pneumophila. Using chimeric molecules in which portions of the zebrafish Tlr4 proteins were fused to portions of the mouse TLR4 protein, we show that the lack of responsiveness to LPS was most likely due to the inability of the extracellular portions of zebrafish Tlr4a and Tlr4b to recognize the molecule, rather than to changes in their capacities to transduce signals through their Toll/IL-1 receptor (TIR) domains. Taken together, these findings strongly support the notion that zebrafish tlr4a and tlr4b paralogs have evolved to provide alternative ligand specificities to the Tlr immune defense system in this species. These data demonstrate that intensive examination of gene histories when describing the Tlr proteins of basally diverging vertebrates is required to obtain fuller appreciation of the evolution of their function. These studies provide the first evidence for the functional evolution of a novel Tlr.


Assuntos
Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/imunologia , Peixe-Zebra/genética , Peixe-Zebra/imunologia , Sequência de Aminoácidos , Animais , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Galinhas , Humanos , Ligantes , Lipopolissacarídeos/fisiologia , Camundongos , Dados de Sequência Molecular , Filogenia , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Isoformas de Proteínas/metabolismo , Homologia de Sequência de Aminoácidos , Receptor 4 Toll-Like/metabolismo , Receptor 4 Toll-Like/fisiologia , Peixe-Zebra/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/fisiologia
16.
Fish Shellfish Immunol ; 25(4): 341-50, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18640057

RESUMO

The zebrafish, Danio rerio, has come to the forefront of biomedical research as a powerful model for the study of development, neurobiology, and genetics of humans. In recent years, use of the zebrafish system has extended into studies in behaviour, immunology and toxicology, retaining the concept that it will serve as a model for human disease. As one of the most thoroughly studied teleosts, with a wealth of genetic and genomic information available, the zebrafish is now being considered as a model for pathogen studies in finfishes. Its genome is currently being sequenced and annotated, and gene microarrays and insertional mutants are commercially available. The use of gene-specific knockdown of translation through morpholino oligonucleotides is widespread. As a result, several laboratories have developed bacterial and viral disease models with the zebrafish to study immune responses to infection. Although many of the zebrafish pathogen models were developed to address human infectious disease, the results of these studies should provide important clues for the development of effective vaccines and prophylactic measures against bacterial and viral pathogens in economically important fishes. In this review, the capabilities and potential of the zebrafish model system will be discussed and an overview of information on zebrafish infectious disease models will be presented.


Assuntos
Infecções Bacterianas/imunologia , Doenças dos Peixes , Viroses/imunologia , Peixe-Zebra/imunologia , Animais , Modelos Animais de Doenças , Doenças dos Peixes/imunologia , Doenças dos Peixes/microbiologia , Doenças dos Peixes/virologia , Técnicas de Silenciamento de Genes
17.
Toxicol Sci ; 98(1): 118-24, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17400579

RESUMO

Arsenic has been associated with a multitude of human health problems; however, its impact on host resistance to infection has not been extensively researched. In vertebrates, the innate immune response is vital for potentiating the adaptive immune response. Therefore, dampening of the innate immune response results in an immunocompromised host. In this present study, effects of low concentrations of arsenic on zebrafish resistance to infection are evaluated. Exposure to 2 and 10 ppb arsenic, both considered safe levels in drinking water, resulted in a greater than 50-fold increase in viral load and at least a 17-fold increase in bacterial load in embryos. To determine the cause of this amplified pathogen load, important components of the innate immune system were analyzed. Presence of arsenic dampened the overall innate immune health of the fish as evidenced by reductions in respiratory burst activity. Viral infection, after arsenic exposure, showed decreases of up to 13- and 1.5-fold changes in interferon and Mx mRNA expression, respectively. Bacterial infection, post arsenic exposure, demonstrated at least 2.5- and 4-fold declines in interleukin-1beta and tumor necrosis factor-alpha mRNA levels, respectively. Maximum expression of these essential cytokines was also delayed upon arsenic exposure. Our data indicate that arsenic exposure, at concentrations deemed safe in drinking water, suppresses the overall innate immune function in zebrafish and present the zebrafish as a unique model for studying immunotoxicity of environmental toxicants. To our knowledge, this is the first report describing the effects of such low levels of arsenic on host resistance to infection.


Assuntos
Arsênio/toxicidade , Imunidade Inata/efeitos dos fármacos , Peixe-Zebra/imunologia , Animais , Atividade Bactericida do Sangue/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias , Citocinas/biossíntese , DNA Complementar/biossíntese , RNA/biossíntese , RNA Mensageiro/biossíntese , Explosão Respiratória/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Necrose Tumoral alfa/biossíntese
18.
J Immunol ; 178(7): 4517-27, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17372010

RESUMO

In mammals, Toll-IL-1R-containing adaptor molecule 1 (TICAM1)-dependent TLR pathways induce NF-kappaB and IFN-beta responses. TICAM1 activates NF-kappaB through two different pathways involving its interactions with TNFR-associated factor 6 and receptor-interacting protein 1. It also activates IFN regulatory factor 3/7 through its interaction with TANK-binding kinase-1, leading to the robust up-regulation of IFN-beta. In this study, we describe the role of zebrafish (Danio rerio) TICAM1 in activating NF-kappaB and zebrafish type I IFN. Zebrafish IFN is unique in that it cannot be categorized as being alpha- or beta-like. Through comprehensive sequence, phylogenetic, and syntenic analyses, we fully describe the identification of a zebrafish TICAM1 ortholog. Zebrafish TICAM1 exhibits sequence divergence from its mammalian orthologs and our data demonstrate that these sequence differences have functional consequences. Zebrafish TICAM1 activates zebrafish IFN; however, it does so in an apparently IFN regulatory factor 3/7-independent manner. Furthermore, zebrafish TICAM1 does not interact with zebrafish TNFR-associated factor 6, thus NF-kappaB activation is dependent upon its interaction with receptor-interacting protein 1. Comparative genome analysis suggests that TICAM1 and TICAM2 evolved from a common vertebrate TICAM ancestor following a gene duplication event and that TICAM2 was lost in teleosts following the divergence of the rayfin and lobefin fishes 450 million years ago. These studies provide evidence, for the first time, of the evolving function of a vertebrate TLR pathway.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/classificação , Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Interferon Tipo I/genética , NF-kappa B/metabolismo , Ativação Transcricional , Peixe-Zebra/imunologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Sequência de Aminoácidos , Animais , Células Cultivadas , Duplicação Gênica , Humanos , Dados de Sequência Molecular , Filogenia , Alinhamento de Sequência , Fator 6 Associado a Receptor de TNF/metabolismo , Receptor 3 Toll-Like/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
19.
Integr Comp Biol ; 46(6): 1040-54, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21672806

RESUMO

Understanding the ecotoxicological effects of arsenic in the environment is paramount to mitigating its deleterious effects on ecological and human health, particularly on the immune response. Toxicological and long-term health effects of arsenic exposure have been well studied. Its specific effects on immune function, however, are less well understood. Eukaryotic immune function often includes both general (innate) as well as specific (adaptive) responses to pathogens. Innate immunity is thought to be the primary defense during early embryonic development, subsequently potentiating adaptive immunity in jawed vertebrates, whereas all other eukaryotes must rely solely on the innate immune response throughout their life cycle. Here, we review the known ecotoxicological effects of arsenic on general health, including immune function, and propose the adoption of zebrafish as a vertebrate model for studying such effects on innate immunity.

20.
Mar Biotechnol (NY) ; 7(5): 494-505, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16007375

RESUMO

The innate immune response, the first line of defense against invading pathogens, can be perturbed by environmental toxicants such as arsenic. This study reports the effects of arsenic on innate immunity of zebrafish. Respiratory burst activity, messenger RNA expression of tumor necrosis factor alpha (TNF-alpha), a primer of the respiratory burst response, and mRNA expression of the antiviral cytokines interferon (IFN) and MX, : before and after viral infection, were examined in arsenic-exposed zebrafish larvae. Respiratory burst activity and TNF-alpha expression were decreased upon arsenic exposure, indicating inhibition of TNF-alpha priming of the respiratory burst response. Arsenic enhanced IFN expression slightly over time, but reduced MX : expression. In zebrafish infected with snakehead rhabdovirus, arsenic decreased induction and altered the kinetics of IFN and MX : upon infection. Differences in IFN and MX : expression in arsenic-exposed larvae point toward an interruption of the Janus kinase-signal transducer and activator of transcription (JAK/STAT) pathway.


Assuntos
Arsênio/toxicidade , Imunidade Inata/efeitos dos fármacos , Explosão Respiratória/efeitos dos fármacos , Peixe-Zebra/imunologia , Peixe-Zebra/metabolismo , Animais , Arsênio/antagonistas & inibidores , Primers do DNA , Infecções por Vírus de DNA/metabolismo , Doenças dos Peixes/imunologia , Expressão Gênica/efeitos dos fármacos , Imunocompetência/efeitos dos fármacos , Interferons/biossíntese , Interferons/genética , Larva/metabolismo , Novirhabdovirus/imunologia , Reação em Cadeia da Polimerase/métodos , RNA Mensageiro/biossíntese , RNA Mensageiro/efeitos dos fármacos , Infecções por Rhabdoviridae/imunologia , Infecções por Rhabdoviridae/veterinária , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética , Peixe-Zebra/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...