Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
PLoS Pathog ; 13(6): e1006467, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28650995

RESUMO

Shigella flexneri, a Gram-negative enteroinvasive pathogen, causes inflammatory destruction of the human intestinal epithelium. Infection by S. flexneri has been well-studied in vitro and is a paradigm for bacterial interactions with the host immune system. Recent work has revealed that components of the cytoskeleton have important functions in innate immunity and inflammation control. Septins, highly conserved cytoskeletal proteins, have emerged as key players in innate immunity to bacterial infection, yet septin function in vivo is poorly understood. Here, we use S. flexneri infection of zebrafish (Danio rerio) larvae to study in vivo the role of septins in inflammation and infection control. We found that depletion of Sept15 or Sept7b, zebrafish orthologs of human SEPT7, significantly increased host susceptibility to bacterial infection. Live-cell imaging of Sept15-depleted larvae revealed increasing bacterial burdens and a failure of neutrophils to control infection. Strikingly, Sept15-depleted larvae present significantly increased activity of Caspase-1 and more cell death upon S. flexneri infection. Dampening of the inflammatory response with anakinra, an antagonist of interleukin-1 receptor (IL-1R), counteracts Sept15 deficiency in vivo by protecting zebrafish from hyper-inflammation and S. flexneri infection. These findings highlight a new role for septins in host defence against bacterial infection, and suggest that septin dysfunction may be an underlying factor in cases of hyper-inflammation.


Assuntos
Disenteria Bacilar/imunologia , Imunidade Inata/imunologia , Septinas/metabolismo , Animais , Modelos Animais de Doenças , Disenteria Bacilar/microbiologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Inflamação/imunologia , Inflamação/microbiologia , Mucosa Intestinal/microbiologia , Larva/metabolismo , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Shigella flexneri , Peixe-Zebra
2.
EMBO Rep ; 18(2): 303-318, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28039206

RESUMO

During infection, plasma membrane (PM) blebs protect host cells against bacterial pore-forming toxins (PFTs), but were also proposed to promote pathogen dissemination. However, the details and impact of blebbing regulation during infection remained unclear. Here, we identify the endoplasmic reticulum chaperone Gp96 as a novel regulator of PFT-induced blebbing. Gp96 interacts with non-muscle myosin heavy chain IIA (NMHCIIA) and controls its activity and remodelling, which is required for appropriate coordination of bleb formation and retraction. This mechanism involves NMHCIIA-Gp96 interaction and their recruitment to PM blebs and strongly resembles retraction of uropod-like structures from polarized migrating cells, a process that also promotes NMHCIIA-Gp96 association. Consistently, Gp96 and NMHCIIA not only protect the PM integrity from listeriolysin O (LLO) during infection by Listeria monocytogenes but also affect cytoskeletal organization and cell migration. Finally, we validate the association between Gp96 and NMHCIIA in vivo and show that Gp96 is required to protect hosts from LLO-dependent killing.


Assuntos
Actomiosina/metabolismo , Toxinas Bacterianas/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Sobrevivência Celular , Humanos , Listeria monocytogenes , Camundongos , Chaperonas Moleculares/metabolismo , Peixe-Zebra
3.
PLoS Pathog ; 9(9): e1003588, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24039575

RESUMO

Autophagy, an ancient and highly conserved intracellular degradation process, is viewed as a critical component of innate immunity because of its ability to deliver cytosolic bacteria to the lysosome. However, the role of bacterial autophagy in vivo remains poorly understood. The zebrafish (Danio rerio) has emerged as a vertebrate model for the study of infections because it is optically accessible at the larval stages when the innate immune system is already functional. Here, we have characterized the susceptibility of zebrafish larvae to Shigella flexneri, a paradigm for bacterial autophagy, and have used this model to study Shigella-phagocyte interactions in vivo. Depending on the dose, S. flexneri injected in zebrafish larvae were either cleared in a few days or resulted in a progressive and ultimately fatal infection. Using high resolution live imaging, we found that S. flexneri were rapidly engulfed by macrophages and neutrophils; moreover we discovered a scavenger role for neutrophils in eliminating infected dead macrophages and non-immune cell types that failed to control Shigella infection. We observed that intracellular S. flexneri could escape to the cytosol, induce septin caging and be targeted to autophagy in vivo. Depletion of p62 (sequestosome 1 or SQSTM1), an adaptor protein critical for bacterial autophagy in vitro, significantly increased bacterial burden and host susceptibility to infection. These results show the zebrafish larva as a new model for the study of S. flexneri interaction with phagocytes, and the manipulation of autophagy for anti-bacterial therapy in vivo.


Assuntos
Autofagia , Disenteria Bacilar/metabolismo , Macrófagos/metabolismo , Neutrófilos/metabolismo , Shigella flexneri/metabolismo , Peixe-Zebra/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Modelos Animais de Doenças , Disenteria Bacilar/genética , Disenteria Bacilar/patologia , Humanos , Macrófagos/microbiologia , Macrófagos/patologia , Neutrófilos/microbiologia , Neutrófilos/patologia , Peixe-Zebra/genética , Peixe-Zebra/microbiologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
4.
Dis Model Mech ; 17(1)2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38131137

RESUMO

Shigella flexneri is a human-adapted pathovar of Escherichia coli that can invade the intestinal epithelium, causing inflammation and bacillary dysentery. Although an important human pathogen, the host response to S. flexneri has not been fully described. Zebrafish larvae represent a valuable model for studying human infections in vivo. Here, we use a Shigella-zebrafish infection model to generate mRNA expression profiles of host response to Shigella infection at the whole-animal level. Immune response-related processes dominate the signature of early Shigella infection (6 h post-infection). Consistent with its clearance from the host, the signature of late Shigella infection (24 h post-infection) is significantly changed, and only a small set of immune-related genes remain differentially expressed, including acod1 and gpr84. Using mutant lines generated by ENU, CRISPR mutagenesis and F0 crispants, we show that acod1- and gpr84-deficient larvae are more susceptible to Shigella infection. Together, these results highlight the power of zebrafish to model infection by bacterial pathogens and reveal the mRNA expression of the early (acutely infected) and late (clearing) host response to Shigella infection.


Assuntos
Disenteria Bacilar , Animais , Humanos , Disenteria Bacilar/genética , Shigella flexneri/genética , Shigella flexneri/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/microbiologia , Inflamação/microbiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
5.
Dis Model Mech ; 13(7)2020 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-32461265

RESUMO

Toxoplasma gondii is an obligate intracellular parasite capable of invading any nucleated cell. Three main clonal lineages (type I, II, III) exist and murine models have driven the understanding of general and strain-specific immune mechanisms underlying Toxoplasma infection. However, murine models are limited for studying parasite-leukocyte interactions in vivo, and discrepancies exist between cellular immune responses observed in mouse versus human cells. Here, we developed a zebrafish infection model to study the innate immune response to Toxoplasma in vivo By infecting the zebrafish hindbrain ventricle, and using high-resolution microscopy techniques coupled with computer vision-driven automated image analysis, we reveal that Toxoplasma invades brain cells and replicates inside a parasitophorous vacuole to which type I and III parasites recruit host cell mitochondria. We also show that type II and III strains maintain a higher infectious burden than type I strains. To understand how parasites are cleared in vivo, we further analyzed Toxoplasma-macrophage interactions using time-lapse microscopy and three-dimensional correlative light and electron microscopy (3D CLEM). Time-lapse microscopy revealed that macrophages are recruited to the infection site and play a key role in Toxoplasma control. High-resolution 3D CLEM revealed parasitophorous vacuole breakage in brain cells and macrophages in vivo, suggesting that cell-intrinsic mechanisms may be used to destroy the intracellular niche of tachyzoites. Together, our results demonstrate in vivo control of Toxoplasma by macrophages, and highlight the possibility that zebrafish may be further exploited as a novel model system for discoveries within the field of parasite immunity.This article has an associated First Person interview with the first author of the paper.


Assuntos
Macrófagos/parasitologia , Rombencéfalo/microbiologia , Toxoplasma/crescimento & desenvolvimento , Toxoplasmose Animal/parasitologia , Toxoplasmose Cerebral/parasitologia , Peixe-Zebra/parasitologia , Animais , Modelos Animais de Doenças , Interações Hospedeiro-Parasita , Macrófagos/imunologia , Macrófagos/ultraestrutura , Microscopia Confocal , Microscopia Eletrônica de Varredura , Microscopia de Fluorescência , Microscopia de Vídeo , Carga Parasitária , Rombencéfalo/imunologia , Rombencéfalo/ultraestrutura , Fatores de Tempo , Toxoplasma/imunologia , Toxoplasma/ultraestrutura , Toxoplasmose Animal/imunologia , Toxoplasmose Animal/patologia , Toxoplasmose Cerebral/imunologia , Toxoplasmose Cerebral/patologia
6.
mBio ; 9(3)2018 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-29946048

RESUMO

Emergency granulopoiesis is a hematopoietic program of stem cell-driven neutrophil production used to counteract immune cell exhaustion following infection. Shigella flexneri is a Gram-negative enteroinvasive pathogen controlled by neutrophils. In this study, we use a Shigella-zebrafish (Danio rerio) infection model to investigate emergency granulopoiesis in vivo We show that stem cell-driven neutrophil production occurs in response to Shigella infection and requires macrophage-independent signaling by granulocyte colony-stimulating factor (Gcsf). To test whether emergency granulopoiesis can function beyond homoeostasis to enhance innate immunity, we developed a reinfection assay using zebrafish larvae that have not yet developed an adaptive immune system. Strikingly, larvae primed with a sublethal dose of Shigella are protected against a secondary lethal dose of Shigella in a type III secretion system (T3SS)-dependent manner. Collectively, these results highlight a new role for emergency granulopoiesis in boosting host defense and demonstrate that zebrafish larvae can be a valuable in vivo model to investigate innate immune memory.IMPORTANCEShigella is an important human pathogen of the gut. Emergency granulopoiesis is the enhanced production of neutrophils by hematopoietic stem and progenitor cells (HSPCs) upon infection and is widely considered a homoeostatic mechanism for replacing exhausted leukocytes. In this study, we developed a Shigella-zebrafish infection model to investigate stem cell-driven emergency granulopoiesis. We discovered that zebrafish initiate granulopoiesis in response to Shigella infection, via macrophage-independent signaling of granulocyte colony-stimulating factor (Gcsf). Strikingly, larvae primed with a sublethal dose of Shigella are protected against a secondary lethal dose of Shigella in a type III secretion system (T3SS)-dependent manner. Taken together, we show that zebrafish infection can be used to capture Shigella-mediated stem cell-driven granulopoiesis and provide a new model system to study stem cell biology in vivo Our results also highlight the potential of manipulating stem cell-driven granulopoiesis to boost innate immunity and combat infectious disease.


Assuntos
Coinfecção/imunologia , Modelos Animais de Doenças , Disenteria Bacilar/microbiologia , Leucopoese , Neutrófilos/imunologia , Shigella flexneri/fisiologia , Animais , Coinfecção/microbiologia , Coinfecção/fisiopatologia , Disenteria Bacilar/imunologia , Disenteria Bacilar/fisiopatologia , Feminino , Humanos , Larva/imunologia , Larva/microbiologia , Macrófagos/imunologia , Masculino , Neutrófilos/citologia , Peixe-Zebra/imunologia , Peixe-Zebra/microbiologia
7.
mBio ; 9(2)2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29588406

RESUMO

Mucormycosis is an emerging fungal infection with extremely high mortality rates in patients with defects in their innate immune response, specifically in functions mediated through phagocytes. However, we currently have a limited understanding of the molecular and cellular interactions between these innate immune effectors and mucormycete spores during the early immune response. Here, the early events of innate immune recruitment in response to infection by Mucor circinelloides spores are modeled by a combined in silico modeling approach and real-time in vivo microscopy. Phagocytes are rapidly recruited to the site of infection in a zebrafish larval model of mucormycosis. This robust early recruitment protects from disease onset in vivoIn silico analysis identified that protection is dependent on the number of phagocytes at the infection site, but not the speed of recruitment. The mathematical model highlights the role of proinflammatory signals for phagocyte recruitment and the importance of inhibition of spore germination for protection from active fungal disease. These in silico data are supported by an in vivo lack of fungal spore killing and lack of reactive oxygen burst, which together result in latent fungal infection. During this latent stage of infection, spores are controlled in innate granulomas in vivo Disease can be reactivated by immunosuppression. Together, these data represent the first in vivo real-time analysis of innate granuloma formation during the early stages of a fungal infection. The results highlight a potential latent stage during mucormycosis that should urgently be considered for clinical management of patients.IMPORTANCE Mucormycosis is a dramatic fungal infection frequently leading to the death of patients. We know little about the immune response to the fungus causing this infection, although evidence points toward defects in early immune events after infection. Here, we dissect this early immune response to infectious fungal spores. We show that specialized white blood cells (phagocytes) rapidly respond to these spores and accumulate around the fungus. However, we demonstrate that the mechanisms that enable phagocytes to kill the fungus fail, allowing for survival of spores. Instead a cluster of phagocytes resembling an early granuloma is formed around spores to control the latent infection. This study is the first detailed analysis of early granuloma formation during a fungal infection highlighting a latent stage that needs to be considered for clinical management of patients.


Assuntos
Granuloma/imunologia , Granuloma/microbiologia , Imunidade Inata/fisiologia , Mucor/patogenicidade , Fagócitos/citologia , Animais , Dexametasona/farmacologia , Interações Hospedeiro-Patógeno , Modelos Teóricos , Neutrófilos/metabolismo , Fagócitos/efeitos dos fármacos , Peixe-Zebra
8.
Nat Microbiol ; 2: 17027, 2017 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-28263305

RESUMO

Pseudomonas aeruginosa is a Gram-negative bacterial pathogen associated with acute and chronic infections. The universal cyclic-di-GMP second messenger is instrumental in the switch from a motile lifestyle to resilient biofilm as in the cystic fibrosis lung. The SadC diguanylate cyclase is associated with this patho-adaptive transition. Here, we identify an unrecognized SadC partner, WarA, which we show is a methyltransferase in complex with a putative kinase, WarB. We established that WarA binds to cyclic-di-GMP, which potentiates its methyltransferase activity. Together, WarA and WarB have structural similarities with the bifunctional Escherichia coli lipopolysaccharide (LPS) O antigen regulator WbdD. Strikingly, WarA influences P. aeruginosa O antigen modal distribution and interacts with the LPS biogenesis machinery. LPS is known to modulate the immune response in the host, and by using a zebrafish infection model, we implicate WarA in the ability of P. aeruginosa to evade detection by the host.


Assuntos
GMP Cíclico/análogos & derivados , Evasão da Resposta Imune , Lipopolissacarídeos/metabolismo , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/patogenicidade , Animais , GMP Cíclico/metabolismo , Modelos Animais de Doenças , Metiltransferases/metabolismo , Ligação Proteica , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/patologia , Peixe-Zebra
9.
Nat Commun ; 8: 15048, 2017 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-28425465

RESUMO

Aquatic chytrid fungi threaten amphibian biodiversity worldwide owing to their ability to rapidly expand their geographical distributions and to infect a wide range of hosts. Combating this risk requires an understanding of chytrid host range to identify potential reservoirs of infection and to safeguard uninfected regions through enhanced biosecurity. Here we extend our knowledge on the host range of the chytrid Batrachochytrium dendrobatidis by demonstrating infection of a non-amphibian vertebrate host, the zebrafish. We observe dose-dependent mortality and show that chytrid can infect and proliferate on zebrafish tissue. We also show that infection phenotypes (fin erosion, cell apoptosis and muscle degeneration) are direct symptoms of infection. Successful infection is dependent on disrupting the zebrafish microbiome, highlighting that, as is widely found in amphibians, commensal bacteria confer protection against this pathogen. Collectively, our findings greatly expand the limited tool kit available to study pathogenesis and host response to chytrid infection.


Assuntos
Anfíbios/microbiologia , Quitridiomicetos/fisiologia , Doenças dos Peixes/microbiologia , Peixe-Zebra/microbiologia , Nadadeiras de Animais/microbiologia , Animais , Interações Hospedeiro-Patógeno , Interações Microbianas , Microbiota/fisiologia
10.
Curr Biol ; 26(24): 3343-3351, 2016 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-27889262

RESUMO

Bdellovibrio bacteriovorus are predatory bacteria that invade and kill a range of Gram-negative bacterial pathogens in natural environments and in vitro [1, 2]. In this study, we investigated Bdellovibrio as an injected, antibacterial treatment in vivo, using zebrafish (Danio rerio) larvae infected with an antibiotic-resistant strain of the human pathogen Shigella flexneri. When injected alone, Bdellovibrio can persist for more than 24 hr in vivo yet exert no pathogenic effects on zebrafish larvae. Bdellovibrio injection of zebrafish containing a lethal dose of Shigella promotes pathogen killing, leading to increased zebrafish survival. Live-cell imaging of infected zebrafish reveals that Shigella undergo rounding induced by the invasive predation from Bdellovibrio in vivo. Furthermore, Shigella-dependent replication of Bdellovibrio was captured inside the zebrafish larvae, indicating active predation in vivo. Bdellovibrio can be engulfed and ultimately eliminated by host neutrophils and macrophages, yet have a sufficient dwell time to prey on pathogens. Experiments in immune-compromised zebrafish reveal that maximal therapeutic benefits of Bdellovibrio result from the synergy of both bacterial predation and host immunity, but that in vivo predation contributes significantly to the survival outcome. Our results demonstrate that successful antibacterial therapy can be achieved via the host immune system working together with bacterial predation by Bdellovibrio. Such cooperation may be important to consider in the fight against antibiotic-resistant infections in vivo.


Assuntos
Antibiose , Bdellovibrio/fisiologia , Disenteria Bacilar/imunologia , Disenteria Bacilar/microbiologia , Shigella flexneri/fisiologia , Animais , Imunidade Celular , Imunidade Inata , Larva/imunologia , Larva/microbiologia , Peixe-Zebra
11.
EMBO Mol Med ; 7(3): 240-58, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25637383

RESUMO

Transplant recipients on calcineurin inhibitors are at high risk of invasive fungal infection. Understanding how calcineurin inhibitors impair fungal immunity is a key priority for defining risk of infection. Here, we show that the calcineurin inhibitor tacrolimus impairs clearance of the major mould pathogen Aspergillus fumigatus from the airway, by inhibiting macrophage inflammatory responses. This leads to defective early neutrophil recruitment and fungal clearance. We confirm these findings in zebrafish, showing an evolutionarily conserved role for calcineurin signalling in neutrophil recruitment during inflammation. We find that calcineurin-NFAT activation is phagocytosis dependent and collaborates with NF-κB for TNF-α production. For yeast zymosan particles, activation of macrophage calcineurin-NFAT occurs via the phagocytic Dectin-1-spleen tyrosine kinase pathway, but for A. fumigatus, activation occurs via a phagosomal TLR9-dependent and Bruton's tyrosine kinase-dependent signalling pathway that is independent of MyD88. We confirm the collaboration between NFAT and NF-κB for TNF-α production in primary alveolar macrophages. These observations identify inhibition of a newly discovered macrophage TLR9-BTK-calcineurin-NFAT signalling pathway as a key immune defect that leads to organ transplant-related invasive aspergillosis.


Assuntos
Aspergillus fumigatus/imunologia , Calcineurina/metabolismo , Macrófagos/imunologia , Fatores de Transcrição NFATC/metabolismo , Fagocitose , Proteínas Tirosina Quinases/metabolismo , Receptor Toll-Like 9/metabolismo , Tirosina Quinase da Agamaglobulinemia , Animais , Aspergilose/imunologia , Aspergilose/microbiologia , Inibidores de Calcineurina/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Imunidade Inata , Macrófagos/microbiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Transdução de Sinais , Tacrolimo/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Peixe-Zebra
12.
J Vis Exp ; (91): e51601, 2014 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-25226510

RESUMO

Shigella flexneri is an intracellular pathogen that can escape from phagosomes to reach the cytosol, and polymerize the host actin cytoskeleton to promote its motility and dissemination. New work has shown that proteins involved in actin-based motility are also linked to autophagy, an intracellular degradation process crucial for cell autonomous immunity. Strikingly, host cells may prevent actin-based motility of S. flexneri by compartmentalizing bacteria inside 'septin cages' and targeting them to autophagy. These observations indicate that a more complete understanding of septins, a family of filamentous GTP-binding proteins, will provide new insights into the process of autophagy. This report describes protocols to monitor autophagy-cytoskeleton interactions caused by S. flexneri in vitro using tissue culture cells and in vivo using zebrafish larvae. These protocols enable investigation of intracellular mechanisms that control bacterial dissemination at the molecular, cellular, and whole organism level.


Assuntos
Disenteria Bacilar/microbiologia , Shigella flexneri/citologia , Animais , Autofagia/fisiologia , Citoesqueleto/microbiologia , Citoesqueleto/patologia , Disenteria Bacilar/patologia , Feminino , Masculino , Shigella flexneri/patogenicidade , Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA