Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cells ; 12(6)2023 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-36980169

RESUMO

Damage-Regulated Autophagy Modulator 1 (DRAM1) is an infection-inducible membrane protein, whose function in the immune response is incompletely understood. Based on previous results in a zebrafish infection model, we have proposed that DRAM1 is a host resistance factor against intracellular mycobacterial infection. To gain insight into the cellular processes underlying DRAM1-mediated host defence, here we studied the interaction of DRAM1 with Mycobacterium marinum in murine RAW264.7 macrophages. We found that, shortly after phagocytosis, DRAM1 localised in a punctate pattern to mycobacteria, which gradually progressed to full DRAM1 envelopment of the bacteria. Within the same time frame, DRAM1-positive mycobacteria colocalised with the LC3 marker for autophagosomes and LysoTracker and LAMP1 markers for (endo)lysosomes. Knockdown analysis revealed that DRAM1 is required for the recruitment of LC3 and for the acidification of mycobacteria-containing vesicles. A reduction in the presence of LAMP1 further suggested reduced fusion of lysosomes with mycobacteria-containing vesicles. Finally, we show that DRAM1 knockdown impairs the ability of macrophages to defend against mycobacterial infection. Together, these results support that DRAM1 promotes the trafficking of mycobacteria through the degradative (auto)phagolysosomal pathway. Considering its prominent effect on host resistance to intracellular infection, DRAM1 is a promising target for therapeutic modulation of the microbicidal capacity of macrophages.


Assuntos
Proteínas de Membrana , Infecções por Mycobacterium , Mycobacterium marinum , Animais , Camundongos , Autofagia , Lisossomos/metabolismo , Macrófagos/metabolismo , Proteínas de Membrana/metabolismo
2.
mBio ; 14(1): e0302422, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36475748

RESUMO

The global burden of tuberculosis (TB) is aggravated by the continuously increasing emergence of drug resistance, highlighting the need for innovative therapeutic options. The concept of host-directed therapy (HDT) as adjunctive to classical antibacterial therapy with antibiotics represents a novel and promising approach for treating TB. Here, we have focused on repurposing the clinically used anticancer drug tamoxifen, which was identified as a molecule with strong host-directed activity against intracellular Mycobacterium tuberculosis (Mtb). Using a primary human macrophage Mtb infection model, we demonstrate the potential of tamoxifen against drug-sensitive as well as drug-resistant Mtb bacteria. The therapeutic effect of tamoxifen was confirmed in an in vivo TB model based on Mycobacterium marinum infection of zebrafish larvae. Tamoxifen had no direct antimicrobial effects at the concentrations used, confirming that tamoxifen acted as an HDT drug. Furthermore, we demonstrate that the antimycobacterial effect of tamoxifen is independent of its well-known target the estrogen receptor (ER) pathway, but instead acts by modulating autophagy, in particular the lysosomal pathway. Through RNA sequencing and microscopic colocalization studies, we show that tamoxifen stimulates lysosomal activation and increases the localization of mycobacteria in lysosomes both in vitro and in vivo, while inhibition of lysosomal activity during tamoxifen treatment partly restores mycobacterial survival. Thus, our work highlights the HDT potential of tamoxifen and proposes it as a repurposed molecule for the treatment of TB. IMPORTANCE Tuberculosis (TB) is the world's most lethal infectious disease caused by a bacterial pathogen, Mycobacterium tuberculosis. This pathogen evades the immune defenses of its host and grows intracellularly in immune cells, particularly inside macrophages. There is an urgent need for novel therapeutic strategies because treatment of TB patients is increasingly complicated by rising antibiotic resistance. In this study, we explored a breast cancer drug, tamoxifen, as a potential anti-TB drug. We show that tamoxifen acts as a so-called host-directed therapeutic, which means that it does not act directly on the bacteria but helps the host macrophages combat the infection more effectively. We confirmed the antimycobacterial effect of tamoxifen in a zebrafish model for TB and showed that it functions by promoting the delivery of mycobacteria to digestive organelles, the lysosomes. These results support the high potential of tamoxifen to be repurposed to fight antibiotic-resistant TB infections by host-directed therapy.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Animais , Humanos , Peixe-Zebra , Tamoxifeno/farmacologia , Tamoxifeno/uso terapêutico , Reposicionamento de Medicamentos , Tuberculose/microbiologia , Antituberculosos/farmacologia , Antituberculosos/uso terapêutico , Mycobacterium tuberculosis/genética
3.
Cell Death Dis ; 11(4): 277, 2020 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-32332700

RESUMO

DNA damage regulated autophagy modulator 1 (DRAM1) is a stress-inducible regulator of autophagy and cell death. DRAM1 has been implicated in cancer, myocardial infarction, and infectious diseases, but the molecular and cellular functions of this transmembrane protein remain poorly understood. Previously, we have proposed DRAM1 as a host resistance factor for tuberculosis (TB) and a potential target for host-directed anti-infective therapies. In this study, we generated a zebrafish dram1 mutant and investigated its loss-of-function effects during Mycobacterium marinum (Mm) infection, a widely used model in TB research. In agreement with previous knockdown analysis, dram1 mutation increased the susceptibility of zebrafish larvae to Mm infection. RNA sequencing revealed major effects of Dram1 deficiency on metabolic, immune response, and cell death pathways during Mm infection, and only minor effects on proteinase and metabolic pathways were found under uninfected conditions. Furthermore, unchallenged dram1 mutants did not display overt autophagic defects, but autophagic targeting of Mm was reduced in the absence of Dram1. The phagocytic ability of macrophages in dram1 mutants was unaffected, but acidification of Mm-containing vesicles was strongly reduced, indicating that Dram1 is required for phagosome maturation. By in vivo imaging, we observed that Dram1-deficient macrophages fail to restrict Mm during early stages of infection. The resulting increase in bacterial burden could be reverted by knockdown of inflammatory caspase a (caspa) and gasdermin Eb (gsdmeb), demonstrating pyroptosis as the mechanism underlying premature cell death of Mm-infected macrophages in dram1 mutants. Collectively, these data demonstrate that dissemination of mycobacterial infection in zebrafish larvae is promoted in the absence of Dram1 due to reduced maturation of mycobacteria-containing vesicles, failed intracellular containment, and consequent pyroptotic death of infected macrophages. These results provide new evidence that Dram1 plays a central role in host resistance to intracellular infection, acting at the crossroad of autophagy and cell death.


Assuntos
Autofagia/genética , Macrófagos/metabolismo , Proteínas de Membrana/deficiência , Infecções por Mycobacterium não Tuberculosas/metabolismo , Piroptose/genética , Tuberculose/genética , Animais , Morte Celular , Humanos , Peixe-Zebra
4.
PLoS Pathog ; 15(2): e1007329, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30818338

RESUMO

Mycobacterial pathogens are the causative agents of chronic infectious diseases like tuberculosis and leprosy. Autophagy has recently emerged as an innate mechanism for defense against these intracellular pathogens. In vitro studies have shown that mycobacteria escaping from phagosomes into the cytosol are ubiquitinated and targeted by selective autophagy receptors. However, there is currently no in vivo evidence for the role of selective autophagy receptors in defense against mycobacteria, and the importance of autophagy in control of mycobacterial diseases remains controversial. Here we have used Mycobacterium marinum (Mm), which causes a tuberculosis-like disease in zebrafish, to investigate the function of two selective autophagy receptors, Optineurin (Optn) and SQSTM1 (p62), in host defense against a mycobacterial pathogen. To visualize the autophagy response to Mm in vivo, optn and p62 zebrafish mutant lines were generated in the background of a GFP-Lc3 autophagy reporter line. We found that loss-of-function mutation of optn or p62 reduces autophagic targeting of Mm, and increases susceptibility of the zebrafish host to Mm infection. Transient knockdown studies confirmed the requirement of both selective autophagy receptors for host resistance against Mm infection. For gain-of-function analysis, we overexpressed optn or p62 by mRNA injection and found this to increase the levels of GFP-Lc3 puncta in association with Mm and to reduce the Mm infection burden. Taken together, our results demonstrate that both Optn and p62 are required for autophagic host defense against mycobacterial infection and support that protection against tuberculosis disease may be achieved by therapeutic strategies that enhance selective autophagy.


Assuntos
Interações Hospedeiro-Patógeno/fisiologia , Infecções por Mycobacterium não Tuberculosas/metabolismo , Mycobacterium marinum/patogenicidade , Animais , Animais Geneticamente Modificados , Autofagia/fisiologia , Proteínas de Ciclo Celular , Modelos Animais de Doenças , Humanos , Macrófagos , Proteínas de Membrana Transportadoras , Mycobacterium/patogenicidade , Infecções por Mycobacterium/metabolismo , Fagossomos , Proteína Sequestossoma-1 , Fator de Transcrição TFIIIA/metabolismo , Tuberculose , Ubiquitina , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
5.
Autophagy ; 15(5): 796-812, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30676840

RESUMO

Innate immune defense against intracellular pathogens, like Salmonella, relies heavily on the autophagy machinery of the host. This response is studied intensively in epithelial cells, the target of Salmonella during gastrointestinal infections. However, little is known of the role that autophagy plays in macrophages, the predominant carriers of this pathogen during systemic disease. Here we utilize a zebrafish embryo model to study the interaction of S. enterica serovar Typhimurium with the macroautophagy/autophagy machinery of macrophages in vivo. We show that phagocytosis of live but not heat-killed Salmonella triggers recruitment of the autophagy marker GFP-Lc3 in a variety of patterns labeling tight or spacious bacteria-containing compartments, also revealed by electron microscopy. Neutrophils display similar GFP-Lc3 associations, but genetic modulation of the neutrophil/macrophage balance and ablation experiments show that macrophages are critical for the defense response. Deficiency of atg5 reduces GFP-Lc3 recruitment and impairs host resistance, in contrast to atg13 deficiency, indicating that Lc3-Salmonella association at this stage is independent of the autophagy preinitiation complex and that macrophages target Salmonella by Lc3-associated phagocytosis (LAP). In agreement, GFP-Lc3 recruitment and host resistance are impaired by deficiency of Rubcn/Rubicon, known as a negative regulator of canonical autophagy and an inducer of LAP. We also found strict dependency on NADPH oxidase, another essential factor for LAP. Both Rubcn and NADPH oxidase are required to activate a Salmonella biosensor for reactive oxygen species inside infected macrophages. These results identify LAP as the major host protective autophagy-related pathway responsible for macrophage defense against Salmonella during systemic infection. Abbreviations: ATG: autophagy related gene; BECN1: Beclin 1; CFU: colony forming units; CYBA/P22PHOX: cytochrome b-245, alpha chain; CYBB/NOX2: cytochrome b-245 beta chain; dpf: days post fertilization; EGFP: enhanced green fluorescent protein; GFP: green fluorescent protein; hfp: hours post fertilization; hpi: hours post infection; IRF8: interferon regulatory factor 8; Lcp1/L-plastin: lymphocyte cytosolic protein 1; LAP: LC3-associated phagocytosis; MAP1LC3/LC3: microtubule-associated protein 1A/1B-light chain 3; mCherry: red fluorescent protein; mpeg1: macrophage expressed gene 1; mpx: myeloid specific peroxidase; NADPH oxidase: nicotinamide adenine dinucleotide phosphate oxidase; NCF4/P40PHOX: neutrophil cytosolic factor 4; NTR-mCherry: nitroreductase-mCherry fusion; PTU: phenylthiourea; PtdIns3K: class III phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol 3-phosphate; RB1CC1/FIP200: RB-1 inducible coiled coin 1; ROS: reactive oxygen species; RT-PCR: reverse transcriptase polymerase chain reaction; RUBCN/RUBICON: RUN and cysteine rich domain containing BECN1-interacting protein; SCV: Salmonella-containing vacuole; S. Typhimurium/S.T: Salmonella enterica serovar Typhimurium; TEM: transmission electron microscopy; Tg: transgenic; TSA: tyramide signal amplification; ULK1/2: unc-51-like autophagy activating kinase 1/2; UVRAG: UVRAG: UV radiation resistance associated; wt: wild type.


Assuntos
Modelos Animais de Doenças , Macrófagos/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia , Fagocitose/genética , Salmonelose Animal , Salmonella typhimurium/imunologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra , Animais , Animais Geneticamente Modificados , Autofagia/fisiologia , Bacteriemia/genética , Bacteriemia/imunologia , Bacteriemia/microbiologia , Bacteriemia/patologia , Embrião não Mamífero , Proteínas Associadas aos Microtúbulos/genética , Fagocitose/imunologia , Espécies Reativas de Oxigênio/metabolismo , Salmonelose Animal/genética , Salmonelose Animal/imunologia , Salmonelose Animal/metabolismo , Salmonelose Animal/microbiologia , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/imunologia , Peixe-Zebra/microbiologia , Proteínas de Peixe-Zebra/genética
6.
Development ; 145(4)2018 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-29475973

RESUMO

Resident microbes promote many aspects of host development, although the mechanisms by which microbiota influence host tissues remain unclear. We showed previously that the microbiota is required for allocation of appropriate numbers of secretory cells in the zebrafish intestinal epithelium. Because Notch signaling is crucial for secretory fate determination, we conducted epistasis experiments to establish whether the microbiota modulates host Notch signaling. We also investigated whether innate immune signaling transduces microbiota cues via the Myd88 adaptor protein. We provide the first evidence that microbiota-induced, Myd88-dependent signaling inhibits host Notch signaling in the intestinal epithelium, thereby promoting secretory cell fate determination. These results connect microbiota activity via innate immune signaling to the Notch pathway, which also plays crucial roles in intestinal homeostasis throughout life and when impaired can result in chronic inflammation and cancer.


Assuntos
Mucosa Intestinal/metabolismo , Microbiota , Fator 88 de Diferenciação Mieloide/metabolismo , Receptores Notch/metabolismo , Animais , Mucosa Intestinal/microbiologia , Mucosa Intestinal/fisiologia , Transdução de Sinais/fisiologia , Peixe-Zebra/metabolismo
7.
PLoS Pathog ; 13(6): e1006437, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28651010

RESUMO

Bacteria of the Burkholderia cepacia complex (Bcc) can cause devastating pulmonary infections in cystic fibrosis (CF) patients, yet the precise mechanisms underlying inflammation, recurrent exacerbations and transition from chronic stages to acute infection and septicemia are not known. Bcc bacteria are generally believed to have a predominant extracellular biofilm life style in infected CF lungs, similar to Pseudomonas aeruginosa, but this has been challenged by clinical observations which show Bcc bacteria predominantly in macrophages. More recently, Bcc bacteria have emerged in nosocomial infections of patients hospitalized for reasons unrelated to CF. Research has abundantly shown that Bcc bacteria can survive and replicate in mammalian cells in vitro, yet the importance of an intracellular life style during infection in humans is unknown. Here we studied the contribution of innate immune cell types to fatal pro-inflammatory infection caused by B. cenocepacia using zebrafish larvae. In strong contrast to the usual protective role for macrophages against microbes, our results show that these phagocytes significantly worsen disease outcome. We provide new insight that macrophages are critical for multiplication of B. cenocepacia in the host and for development of a fatal, pro-inflammatory response that partially depends on Il1-signalling. In contrast, neutrophils did not significantly contribute to disease outcome. In subcutaneous infections that are dominated by neutrophil-driven phagocytosis, the absence of a functional NADPH oxidase complex resulted in a small but measurably higher increase in bacterial growth suggesting the oxidative burst helps limit bacterial multiplication; however, neutrophils were unable to clear the bacteria. We suggest that paradigm-changing approaches are needed for development of novel antimicrobials to efficiently disarm intracellular bacteria of this group of highly persistent, opportunistic pathogens.


Assuntos
Burkholderia cenocepacia/isolamento & purificação , Infecção Hospitalar/microbiologia , Inflamação/microbiologia , Macrófagos/microbiologia , Neutrófilos/microbiologia , Animais , Infecções por Burkholderia/imunologia , Complexo Burkholderia cepacia/imunologia , Fibrose Cística/complicações , Humanos , Pulmão/microbiologia , Neutrófilos/imunologia , Fagocitose/imunologia , Pseudomonas aeruginosa/fisiologia , Infecções Respiratórias/microbiologia
8.
Autophagy ; 10(12): 2389-91, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25484076

RESUMO

Autophagy provides an important defense mechanism against intracellular bacteria, such as Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis disease (TB). We recently reported that pathogen recognition and antibacterial autophagy are connected by the induction of the DNA damage-regulated autophagy modulator DRAM1 via the toll-like receptor (TLR)-MYD88-NFKB innate immunity signaling pathway. Having shown that DRAM1 colocalizes with Mtb in human macrophages, we took advantage of a zebrafish model for TB to investigate the function of DRAM1 in autophagic host defense in vivo. We found that DRAM1 protects the zebrafish host from infection with Mycobacterium marinum (Mm), a close relative of Mtb. Overexpression of DRAM1 increases autophagosome formation and promotes autophagic flux by a mechanism dependent on the cytosolic DNA sensor TMEM173/STING and the ubiquitin receptor SQSTM1/p62. Here we summarize and discuss the implications of these findings.


Assuntos
Autofagia , Macrófagos/microbiologia , Proteínas de Membrana/metabolismo , Infecções por Mycobacterium/metabolismo , Mycobacterium/patogenicidade , Fator 88 de Diferenciação Mieloide/metabolismo , Animais , Humanos
9.
J Virol ; 88(20): 12026-40, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25100833

RESUMO

Hemorrhagic viral diseases are distributed worldwide with important pathogens, such as dengue virus or hantaviruses. The lack of adequate in vivo infection models has limited the research on viral pathogenesis and the current understanding of the underlying infection mechanisms. Although hemorrhages have been associated with the infection of endothelial cells, other cellular types could be the main targets for hemorrhagic viruses. Our objective was to take advantage of the use of zebrafish larvae in the study of viral hemorrhagic diseases, focusing on the interaction between viruses and host cells. Cellular processes, such as transendothelial migration of leukocytes, virus-induced pyroptosis of macrophages. and interleukin-1ß (Il-1ß) release, could be observed in individual cells, providing a deeper knowledge of the immune mechanisms implicated in the disease. Furthermore, the application of these techniques to other pathogens will improve the current knowledge of host-pathogen interactions and increase the potential for the discovery of new therapeutic targets. Importance: Pathogenic mechanisms of hemorrhagic viruses are diverse, and most of the research regarding interactions between viruses and host cells has been performed in cell lines that might not be major targets during natural infections. Thus, viral pathogenesis research has been limited because of the lack of adequate in vivo infection models. The understanding of the relative pathogenic roles of the viral agent and the host response to the infection is crucial. This will be facilitated by the establishment of in vivo infection models using organisms such as zebrafish, which allows the study of the diseases in the context of a complete individual. The use of this animal model with other pathogens could improve the current knowledge on host-pathogen interactions and increase the potential for the discovery of new therapeutic targets against diverse viral diseases.


Assuntos
Apoptose , Interleucina-1beta/metabolismo , Larva/metabolismo , Macrófagos/imunologia , Peixe-Zebra/crescimento & desenvolvimento , Animais , Marcação In Situ das Extremidades Cortadas , Larva/virologia
10.
ACS Nano ; 8(7): 7014-26, 2014 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-24945994

RESUMO

Nanoparticles (NPs) enclosing antibiotics have provided promising therapy against Mycobacterium tuberculosis (Mtb) in different mammalian models. However, the NPs were not visualized in any of these animal studies. Here, we introduce the transparent zebrafish embryo as a system for noninvasive, simultaneous imaging of fluorescent NPs and the fish tuberculosis (TB) agent Mycobacterium marinum (Mm). The study was facilitated by the use of transgenic lines of macrophages, neutrophils, and endothelial cells expressing fluorescent markers readily visible in the live vertebrate. Intravenous injection of Mm led to phagocytosis by blood macrophages. These remained within the vasculature until 3 days postinfection where they started to extravasate and form aggregates of infected cells. Correlative light/electron microscopy revealed that these granuloma-like structures had significant access to the vasculature. Injection of NPs induced rapid uptake by both infected and uninfected macrophages, the latter being actively recruited to the site of infection, thereby providing an efficient targeting into granulomas. Rifampicin-loaded NPs significantly improved embryo survival and lowered bacterial load, as shown by quantitative fluorescence analysis. Our results argue that zebrafish embryos offer a powerful system for monitoring NPs in vivo and rationalize why NP therapy was so effective against Mtb in earlier studies; bacteria and NPs share the same cellular niche.


Assuntos
Portadores de Fármacos/química , Embrião não Mamífero/microbiologia , Mycobacterium marinum/efeitos dos fármacos , Nanopartículas/química , Imagem Óptica , Peixe-Zebra/embriologia , Peixe-Zebra/microbiologia , Animais , Antibacterianos/química , Antibacterianos/farmacologia , Transporte Biológico , Cumarínicos/química , Portadores de Fármacos/metabolismo , Granuloma/microbiologia , Ácido Láctico/química , Macrófagos/microbiologia , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium marinum/fisiologia , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Rodaminas/química , Rifampina/química , Rifampina/farmacologia , Tiazóis/química , Tuberculose/microbiologia , Tuberculose/veterinária
11.
Cell Host Microbe ; 15(6): 753-67, 2014 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-24922577

RESUMO

Autophagy is an important defense mechanism against mycobacteria, the causative agents of tuberculosis. The molecular mechanisms that link mycobacterial recognition to autophagy remain unclear. Our analysis in zebrafish and human macrophage models of mycobacterial infection reveals that the DNA damage-regulated autophagy modulator DRAM1 functions downstream of pathogen recognition by the Toll-like receptor (TLR)/interleukin-1 receptor (IL1R)-MYD88-NF-κB innate immune sensing pathway to activate selective autophagy. Mycobacterial infection of human macrophages and zebrafish embryos induced DRAM1 expression in a MYD88 and NF-κB-dependent manner. DRAM1 knockdown increased mycobacterial infection, whereas overexpression lowered infection by hyperactivating autophagy. DRAM1-mediated selective autophagic defenses require the cytosolic DNA sensor STING and the selective autophagy receptor p62/SQSTM1. Contrary to its known role in autophagy-mediated cell death and cancer, this DRAM1 function is p53 independent. We propose that DRAM1 mediates autophagic defense against a broader range of intracellular pathogens, since DRAM1 expression was also induced by the common bacterial endotoxin lipopolysaccharide.


Assuntos
Autofagia , Macrófagos/microbiologia , Proteínas de Membrana/metabolismo , Infecções por Mycobacterium/metabolismo , Mycobacterium/patogenicidade , Fator 88 de Diferenciação Mieloide/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Autofagia/imunologia , Células Cultivadas , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/microbiologia , Regulação da Expressão Gênica , Genes p53 , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Lipopolissacarídeos/farmacologia , Lisossomos/metabolismo , Macrófagos/fisiologia , Proteínas de Membrana/genética , Infecções por Mycobacterium/imunologia , Infecções por Mycobacterium/microbiologia , NF-kappa B/metabolismo , Receptores de Interleucina-1/metabolismo , Proteína Sequestossoma-1 , Peixe-Zebra/embriologia , Peixe-Zebra/microbiologia
12.
Dis Model Mech ; 6(3): 841-54, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23471913

RESUMO

Toll-like receptors (TLRs) are an important class of pattern recognition receptors (PRRs) that recognize microbial and danger signals. Their downstream signaling upon ligand binding is vital for initiation of the innate immune response. In human and mammalian models, myeloid differentiation factor 88 (MYD88) is known for its central role as an adaptor molecule in interleukin 1 receptor (IL-1R) and TLR signaling. The zebrafish is increasingly used as a complementary model system for disease research and drug screening. Here, we describe a zebrafish line with a truncated version of MyD88 as the first zebrafish mutant for a TLR signaling component. We show that this immune-compromised mutant has a lower survival rate under standard rearing conditions and is more susceptible to challenge with the acute bacterial pathogens Edwardsiella tarda and Salmonella typhimurium. Microarray and quantitative PCR analysis revealed that expression of genes for transcription factors central to innate immunity (including NF-ĸB and AP-1) and the pro-inflammatory cytokine Il1b, is dependent on MyD88 signaling during these bacterial infections. Nevertheless, expression of immune genes independent of MyD88 in the myd88 mutant line was sufficient to limit growth of an attenuated S. typhimurium strain. In the case of infection with the chronic bacterial pathogen Mycobacterium marinum, we show that MyD88 signaling has an important protective role during early pathogenesis. During mycobacterial infection, the myd88 mutant shows accelerated formation of granuloma-like aggregates and increased bacterial burden, with associated lower induction of genes central to innate immunity. This zebrafish myd88 mutant will be a valuable tool for further study of the role of IL1R and TLR signaling in the innate immunity processes underlying infectious diseases, inflammatory disorders and cancer.


Assuntos
Imunidade Inata/genética , Mutação/genética , Fator 88 de Diferenciação Mieloide/genética , Transcrição Gênica , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Peixe-Zebra/imunologia , Sequência de Aminoácidos , Animais , Infecções Bacterianas/genética , Infecções Bacterianas/imunologia , Infecções Bacterianas/microbiologia , Infecções Bacterianas/patologia , Sequência de Bases , Suscetibilidade a Doenças , Edwardsiella tarda/efeitos dos fármacos , Edwardsiella tarda/fisiologia , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/imunologia , Embrião não Mamífero/microbiologia , Embrião não Mamífero/patologia , Flagelina/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Homozigoto , Humanos , Imunidade Inata/efeitos dos fármacos , Leucócitos/efeitos dos fármacos , Leucócitos/metabolismo , Lipopolissacarídeos/farmacologia , Dados de Sequência Molecular , Mycobacterium marinum/efeitos dos fármacos , Mycobacterium marinum/fisiologia , Fator 88 de Diferenciação Mieloide/química , Fator 88 de Diferenciação Mieloide/metabolismo , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia , Poli I-C/farmacologia , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Análise de Sobrevida , Transcrição Gênica/efeitos dos fármacos , Transcriptoma/genética , Peixe-Zebra/microbiologia , Proteínas de Peixe-Zebra/química , Proteínas de Peixe-Zebra/metabolismo
13.
Methods Cell Biol ; 105: 273-308, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21951535

RESUMO

The major cell types of the innate immune system, macrophages and neutrophils, develop during the first two days of zebrafish embryogenesis. The interaction of these immune cells with pathogenic microbes can excellently be traced in the optically transparent zebrafish embryos. Various tools and methods have recently been developed for visualizing and isolating the zebrafish embryonic innate immune cells, for establishing infections by different micro-injection techniques, and for analyzing the host innate immune response following microbial recognition. Here we provide practical guidelines for the application of these methodologies and review the current state of the art in zebrafish infectious disease research.


Assuntos
Biologia do Desenvolvimento/métodos , Técnicas de Silenciamento de Genes/métodos , Genômica/métodos , Imunidade Inata , Hibridização in Situ Fluorescente/métodos , Microinjeções/métodos , Imagem Molecular/métodos , Peixe-Zebra/imunologia , Animais , Biomarcadores/análise , Linhagem da Célula , Embrião não Mamífero , Citometria de Fluxo , Ensaios de Triagem em Larga Escala , Humanos , Macrófagos/imunologia , Neutrófilos/imunologia , Salmonelose Animal/imunologia , Salmonelose Animal/microbiologia , Salmonella typhimurium/imunologia , Transgenes , Peixe-Zebra/embriologia , Peixe-Zebra/genética
14.
Am J Surg ; 195(2): 259-69, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18154764

RESUMO

BACKGROUND: Carotid endarterectomy (CEA) is still considered the "gold-standard" of the treatment of patients with significant carotid stenosis and has proven its value during past decades. However, endovascular techniques have recently been evolving. Carotid artery stenting (CAS) is challenging CEA for the best treatment in patients with carotid stenosis. This review presents the development of CAS according to early reports, results of recent randomized trials, and future perspectives regarding CAS. METHODS: A literature search using the PubMed and Cochrane databases identified articles focusing on the key issues of CEA and CAS. RESULTS: Early nonrandomized reports of CAS showed variable results, and the Stenting and Angioplasty With Protection in Patients at High Risk for Endarterectomy trial led to United States Food and Drug Administration approval of CAS for the treatment of patients with symptomatic carotid stenosis. In contrast, recent trials, such as the Stent-Protected Angioplasty Versus Carotid Endarterectomy trial and the Endarterectomy Versus Stenting in Patients with Symptomatic Severe Carotid Stenosis trial, (re)fuelled the debate between CAS and CEA. In the Stent-Protected Angioplasty Versus Carotid Endarterectomy trial, the complication rate of ipsilateral stroke or death at 30 days was 6.8% for CAS versus 6.3% for CEA and showed that CAS failed the noninferiority test. Analysis of the Endarterectomy Versus Stenting in Patients With Symptomatic Severe Carotid Stenosis trial showed a significant higher risk for death or any stroke at 30 days for endovascular treatment (9.6%) compared with CEA (3.9%). Other aspects-such as evolving best medical treatment, timely intervention, interventionalists' experience, and analysis of plaque composition-may have important influences on the future treatment of patients with carotid artery stenosis. CONCLUSIONS: CAS performed with or without embolic-protection devices can be an effective treatment for patients with carotid artery stenosis. However, presently there is no evidence that CAS provides better results in the prevention of stroke compared with CEA.


Assuntos
Angioplastia com Balão/métodos , Estenose das Carótidas/mortalidade , Estenose das Carótidas/terapia , Endarterectomia das Carótidas/métodos , Stents , Acidente Vascular Cerebral/prevenção & controle , Angioplastia com Balão/efeitos adversos , Estenose das Carótidas/diagnóstico por imagem , Estenose das Carótidas/fisiopatologia , Endarterectomia das Carótidas/efeitos adversos , Feminino , Seguimentos , Humanos , Masculino , Estudos Multicêntricos como Assunto , Complicações Pós-Operatórias/mortalidade , Ensaios Clínicos Controlados Aleatórios como Assunto , Medição de Risco , Sensibilidade e Especificidade , Índice de Gravidade de Doença , Acidente Vascular Cerebral/etiologia , Análise de Sobrevida , Resultado do Tratamento , Ultrassonografia Doppler
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA