Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros













Base de dados
Intervalo de ano de publicação
1.
Cell ; 184(24): 5950-5969.e22, 2021 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-34741801

RESUMO

The biogenesis of mammalian autophagosomes remains to be fully defined. Here, we used cellular and in vitro membrane fusion analyses to show that autophagosomes are formed from a hitherto unappreciated hybrid membrane compartment. The autophagic precursors emerge through fusion of FIP200 vesicles, derived from the cis-Golgi, with endosomally derived ATG16L1 membranes to generate a hybrid pre-autophagosomal structure, HyPAS. A previously unrecognized apparatus defined here controls HyPAS biogenesis and mammalian autophagosomal precursor membranes. HyPAS can be modulated by pharmacological agents whereas its formation is inhibited upon severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or by expression of SARS-CoV-2 nsp6. These findings reveal the origin of mammalian autophagosomal membranes, which emerge via convergence of secretory and endosomal pathways, and show that this process is targeted by microbial factors such as coronaviral membrane-modulating proteins.


Assuntos
Autofagossomos/virologia , COVID-19/virologia , Autofagia , COVID-19/metabolismo , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Retículo Endoplasmático/metabolismo , Endossomos/fisiologia , Endossomos/virologia , Complexo de Golgi/fisiologia , Células HEK293 , Células HeLa , Humanos , Fusão de Membrana , Microscopia Confocal , Fagossomos/metabolismo , Fagossomos/virologia , Proteínas Qa-SNARE/biossíntese , Receptores sigma/biossíntese , SARS-CoV-2 , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/biossíntese , Sinaptotagminas/biossíntese , Receptor Sigma-1
2.
Sci Rep ; 11(1): 5025, 2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33658544

RESUMO

Bioconversion of organic materials is the foundation of many applications in chemical engineering, microbiology and biochemistry. Herein, we introduce a new methodology to quantitatively determine conversion of biomass in viral infections while simultaneously imaging morphological changes of the host cell. As proof of concept, the viral replication of an unidentified giant DNA virus and the cellular response of an amoebal host are studied using soft X-ray microscopy, titration dilution measurements and thermal gravimetric analysis. We find that virions produced inside the cell are visible from 18 h post infection and their numbers increase gradually to a burst size of 280-660 virions. Due to the large size of the virion and its strong X-ray absorption contrast, we estimate that the burst size corresponds to a conversion of 6-12% of carbonaceous biomass from amoebal host to virus. The occurrence of virion production correlates with the appearance of a possible viral factory and morphological changes in the phagosomes and contractile vacuole complex of the amoeba, whereas the nucleus and nucleolus appear unaffected throughout most of the replication cycle.


Assuntos
Acanthamoeba/virologia , Vírus de DNA/ultraestrutura , DNA Viral/genética , Genoma Viral , Vírus Gigantes/ultraestrutura , Vírion/ultraestrutura , Acanthamoeba/ultraestrutura , Biomassa , Vírus de DNA/genética , Vírus de DNA/crescimento & desenvolvimento , Vírus de DNA/isolamento & purificação , DNA Viral/biossíntese , Vírus Gigantes/genética , Vírus Gigantes/crescimento & desenvolvimento , Vírus Gigantes/isolamento & purificação , Interações Hospedeiro-Patógeno/genética , Fagossomos/ultraestrutura , Fagossomos/virologia , Microbiologia do Solo , Termogravimetria , Vacúolos/ultraestrutura , Vacúolos/virologia , Vírion/genética , Vírion/crescimento & desenvolvimento , Replicação Viral , Microtomografia por Raio-X
3.
Trends Microbiol ; 29(6): 528-541, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33243546

RESUMO

Bacteriophages (phages) are the most abundant biological entity in the human body, but until recently the role that phages play in human health was not well characterized. Although phages do not cause infections in human cells, phages can alter the severity of bacterial infections by the dissemination of virulence factors amongst bacterial hosts. Recent studies, made possible with advances in genome engineering and microscopy, have uncovered a novel role for phages in the human body - the ability to modulate the physiology of the mammalian cells that can harbor intracellular bacteria. In this review, we synthesize key results on how phages traverse through mammalian cells - including uptake, distribution, and interaction with intracellular receptors - highlighting how these steps in turn influence host cell killing of bacteria. We discuss the implications of the growing field of phage-mammalian cell interactions for phage therapy.


Assuntos
Bacteriófagos/metabolismo , Células/metabolismo , Células/virologia , Interações Hospedeiro-Patógeno , Mamíferos , Animais , Bacteriófagos/genética , Células/citologia , Citosol/microbiologia , Citosol/virologia , DNA Viral , Humanos , Camundongos , Fagossomos/microbiologia , Fagossomos/virologia , Prófagos/genética , Prófagos/metabolismo , Internalização do Vírus
4.
J Eukaryot Microbiol ; 65(5): 694-704, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29505174

RESUMO

For the first time, ciliates have been found to activate rather than inactivate a virus, chum salmon reovirus (CSV). Activation was seen as an increase in viral titre upon incubation of CSV at 22 °C with Tetrahymena canadenesis and two strains of T. thermophila: wild type (B1975) and a temperature conditional mutant for phagocytosis (NP1). The titre increase was not likely due to replication because CSV had no visible effects on the ciliates and no vertebrate virus has ever been shown unequivocally to replicate in ciliates. When incubated with B1975 and NP1 at 30 °C, CSV was activated only by B1975. Therefore, activation required CSV internalization because at 30 °C only B1975 exhibited phagocytosis. CSV replicated in fish cells at 18 to 26 °C but not at 30 °C. Collectively, these observations point to CSV activation being distinct from replication. Activation is attributed to the CSV capsid being modified in the ciliate phagosomal-lysosomal system and released in a more infectious form. When allowed to swim in CSV-infected fish cell cultures, collected, washed, and transferred to uninfected cultures, T. canadensis caused a CSV infection. Overall the results suggest that ciliates could have roles in the environmental dissemination of some fish viral diseases.


Assuntos
Infecções por Reoviridae/veterinária , Reoviridae/fisiologia , Tetrahymena thermophila/virologia , Animais , Doenças dos Peixes/virologia , Oncorhynchus keta/parasitologia , Oncorhynchus keta/virologia , Fagossomos/virologia , Infecções por Reoviridae/virologia , Temperatura , Ativação Viral , Replicação Viral
5.
J Gen Virol ; 98(3): 486-495, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28008822

RESUMO

Duck enteritis virus (DEV) is an acute, septic, sexually transmitted disease that occurs in ducks, geese and other poultry. Autophagy is an evolutionarily ancient pathway that is important in many viral infections. Despite extensive study, the interplay between DEV and autophagy of host cells is not clearly understood. In this study, we found that DEV infection triggers autophagy in duck embryo fibroblast (DEF) cells, as demonstrated by the appearance of autophagosome-like double- or single-membrane vesicles in the cytoplasm of host cells and the number of GFP-LC3 dots. In addition, increased conversion of the autophagy marker protein LC3-I and LC3-II and decreased p62/SQSTM1 indicated complete autophagy flux. Heat-inactivated DEV infection did not induce autophagy, suggesting that the trigger of autophagy in DEF cells depended on DEV replication. When autophagy was pharmacologically inhibited by LY294002 or wortmannin, DEV replication decreased. The DEV offspring yield decreased when small interference RNA was used to interfere with autophagy related to the genes Beclin-1 and ATG5. In contrast, after treating DEF cells with rapamycin, an inducer of autophagy, DEV replication increased. These results indicated that DEV infection induced autophagy in DEF cells and autophagy facilitated DEV replication.


Assuntos
Autofagia , Mardivirus/fisiologia , Doença de Marek/virologia , Replicação Viral , Androstadienos/farmacologia , Animais , Autofagia/efeitos dos fármacos , Autofagia/genética , Proteína 5 Relacionada à Autofagia/genética , Proteína Beclina-1/genética , Cromonas/farmacologia , Patos , Fibroblastos/virologia , Proteínas Associadas aos Microtúbulos/metabolismo , Morfolinas/farmacologia , Fagossomos/metabolismo , Fagossomos/virologia , RNA Interferente Pequeno , Sirolimo/farmacologia , Wortmanina
6.
Exp Cell Res ; 349(2): 255-263, 2016 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-27793649

RESUMO

Coxsackievirus group B (CVB) is one of the common pathogens that cause myocarditis and cardiomyopathy. Evidence has shown that CVB replication in cardiomyocytes is responsible for the damage and loss of cardiac muscle and the dysfunction of the heart. However, it remains largely undefined how CVB would directly impact cardiac fibroblasts, the most abundant cells in human heart. In this study, cardiac fibroblasts were isolated from Balb/c mice and infected with CVB type 3 (CVB3). Increased double-membraned, autophagosome-like vesicles in the CVB3-infected cardiac fibroblasts were observed with electron microscope. Punctate distribution of LC3 and increased level of LC3-II were also detected in the infected cardiac fibroblasts. Furthermore, we observed that the expression of pro-inflammatory cytokines, IL-6 and TNF-α, was increased in the CVB3-infected cardiac fibroblasts, while suppressed autophagy by 3-MA and Atg7-siRNA inhibited cytokine expression. Consistent with the in vitro findings, increased formation of autophagosomes was observed in the cardiac fibroblasts of Balb/c mice infected with CVB3. In conclusion, our data demonstrated that cardiac fibroblasts respond to CVB3 infection with the formation of autophagosomes and the release of the pro-inflammatory cytokines. These results suggest that the autophagic response of cardiac fibroblasts may play a role in the pathogenesis of myocarditis caused by CVB3 infection.


Assuntos
Autofagossomos/virologia , Enterovirus Humano B , Fibroblastos/virologia , Miocardite/virologia , Miócitos Cardíacos/virologia , Animais , Autofagia/fisiologia , Enterovirus Humano B/fisiologia , Camundongos Endogâmicos BALB C , Miócitos Cardíacos/patologia , Fagossomos/virologia , Replicação Viral/genética
7.
Sci Rep ; 6: 28171, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27302320

RESUMO

To survive and replicate in macrophages Mycobacterium tuberculosis (Mtb) has developed strategies to subvert host defence mechanisms, including autophagy. Autophagy induction has the potential to clear Mtb, but little is known about its effect during controlled tuberculosis and HIV co-infection. Mammalian target of rapamycin complex1 (mTORC1) inhibitors were used to induce autophagy in human macrophages pre-infected with HIV-1BaL and infected with a low dose of Mtb (co-infected), or single Mtb infected (single infected). The controlled Mtb infection was disrupted upon mTOR inhibition resulting in increased Mtb replication in a dose-dependent manner which was more pronounced during co-infection. The increased Mtb replication could be explained by the marked reduction in phagosome acidification upon mTOR inhibition. Autophagy stimulation targeting mTORC1 clearly induced a basal autophagy with flux that was unlinked to the subcellular environment of the Mtb vacuoles, which showed a concurrent suppression in acidification and maturation/flux. Overall our findings indicate that mTOR inhibition during Mtb or HIV/Mtb co-infection interferes with phagosomal maturation, thereby supporting mycobacterial growth during low-dose and controlled infection. Therefore pharmacological induction of autophagy through targeting of the canonical mTORC1-pathway should be handled with caution during controlled tuberculosis, since this could have serious consequences for patients with HIV/Mtb co-infection.


Assuntos
Autofagia/fisiologia , Infecções por HIV/microbiologia , Macrófagos/microbiologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Mycobacterium tuberculosis/patogenicidade , Autofagia/efeitos dos fármacos , Coinfecção , Regulação da Expressão Gênica , Infecções por HIV/genética , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/virologia , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/metabolismo , Naftiridinas/farmacologia , Fagossomos/microbiologia , Fagossomos/virologia , Fosforilação , Proteína Sequestossoma-1/metabolismo , Sirolimo/farmacologia , Tuberculose/metabolismo , Tuberculose/virologia
8.
Autophagy ; 12(9): 1507-20, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27305174

RESUMO

Adenosine monophosphate-activated protein kinase (AMPK) is a crucial energy sensor that maintains cellular energy homeostasis. AMPK plays a critical role in macroautophagy/autophagy, and autophagy facilitates hepatitis B virus (HBV) replication. To date, the intrinsic link among AMPK, autophagy and HBV production remains to be elucidated. Here, we demonstrate that PRKAA (a catalytic subunit of AMPK) is activated in response to HBV-induced oxidative stress, which in turn decreases the production of HBV. Mechanistic studies reveal that the autophagy machinery is associated with the inhibitory effect of PRKAA/AMPK on HBV production. Activation of PRKAA/AMPK promotes autolysosome-dependent degradation through stimulation of cellular ATP levels, which then leads to the depletion of autophagic vacuoles. Taken together, our data suggest that the activation of AMPK might be a stress response of host cells to restrict virus production through promotion of autophagic degradation. These findings therefore indicate that AMPK could provide a potential therapeutic target for HBV infection.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Autofagia/fisiologia , Vírus da Hepatite B/fisiologia , Fagossomos/metabolismo , Replicação Viral , Trifosfato de Adenosina/química , Animais , Replicação do DNA , Ativação Enzimática , Proteínas de Fluorescência Verde/metabolismo , Células Hep G2 , Humanos , Concentração de Íons de Hidrogênio , Lisossomos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Associadas aos Microtúbulos/metabolismo , Oligonucleotídeos/genética , Estresse Oxidativo , Fagossomos/virologia , RNA Interferente Pequeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Vacúolos/metabolismo
9.
Sci Rep ; 6: 23864, 2016 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-27029407

RESUMO

Autophagy is an evolutionarily ancient pathway that has been shown to be important in the innate immune defense against several viruses. However, little is known about the regulatory role of autophagy in transmissible gastroenteritis virus (TGEV) replication. In this study, we found that TGEV infection increased the number of autophagosome-like double- and single-membrane vesicles in the cytoplasm of host cells, a phenomenon that is known to be related to autophagy. In addition, virus replication was required for the increased amount of the autophagosome marker protein LC3-II. Autophagic flux occurred in TGEV-infected cells, suggesting that TGEV infection triggered a complete autophagic response. When autophagy was pharmacologically inhibited by wortmannin or LY294002, TGEV replication increased. The increase in virus yield via autophagy inhibition was further confirmed by the use of siRNA duplexes, through which three proteins required for autophagy were depleted. Furthermore, TGEV replication was inhibited when autophagy was activated by rapamycin. The antiviral response of autophagy was confirmed by using siRNA to reduce the expression of gene p300, which otherwise inhibits autophagy. Together, the results indicate that TGEV infection activates autophagy and that autophagy then inhibits further TGEV replication.


Assuntos
Autofagia/genética , Células Epiteliais/virologia , Interações Hospedeiro-Patógeno , Vírus da Gastroenterite Transmissível/fisiologia , Fatores de Transcrição de p300-CBP/genética , Androstadienos/farmacologia , Animais , Anticorpos Monoclonais/farmacologia , Autofagia/efeitos dos fármacos , Proteína 5 Relacionada à Autofagia/antagonistas & inibidores , Proteína 5 Relacionada à Autofagia/genética , Proteína 5 Relacionada à Autofagia/metabolismo , Proteína 7 Relacionada à Autofagia/antagonistas & inibidores , Proteína 7 Relacionada à Autofagia/genética , Proteína 7 Relacionada à Autofagia/metabolismo , Linhagem Celular , Cromonas/farmacologia , Vesículas Citoplasmáticas/efeitos dos fármacos , Vesículas Citoplasmáticas/metabolismo , Vesículas Citoplasmáticas/virologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Morfolinas/farmacologia , Fagossomos/efeitos dos fármacos , Fagossomos/metabolismo , Fagossomos/virologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Sirolimo/farmacologia , Suínos , Vírus da Gastroenterite Transmissível/patogenicidade , Replicação Viral/efeitos dos fármacos , Wortmanina , Fatores de Transcrição de p300-CBP/antagonistas & inibidores , Fatores de Transcrição de p300-CBP/metabolismo
10.
Adv Virus Res ; 95: 149-95, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27112282

RESUMO

Autophagy is an important cellular catabolic process conserved from yeast to man. Double-membrane vesicles deliver their cargo to the lysosome for degradation. Hence, autophagy is one of the key mechanisms mammalian cells deploy to rid themselves of intracellular pathogens including viruses. However, autophagy serves many more functions during viral infection. First, it regulates the immune response through selective degradation of immune components, thus preventing possibly harmful overactivation and inflammation. Additionally, it delivers virus-derived antigens to antigen-loading compartments for presentation to T lymphocytes. Second, it might take an active part in the viral life cycle by, eg, facilitating its release from cells. Lastly, in the constant arms race between host and virus, autophagy is often hijacked by viruses and manipulated to their own advantage. In this review, we will highlight key steps during viral infection in which autophagy plays a role. We have selected some exemplary viruses and will describe the molecular mechanisms behind their intricate relationship with the autophagic machinery, a result of host-pathogen coevolution.


Assuntos
Autofagia/imunologia , Imunidade Inata , Vírion/imunologia , Viroses/imunologia , Replicação Viral/imunologia , Vírus/imunologia , Imunidade Adaptativa , Animais , Autofagia/genética , Diferenciação Celular , Regulação da Expressão Gênica/imunologia , Interações Hospedeiro-Patógeno , Humanos , Fagossomos/imunologia , Fagossomos/metabolismo , Fagossomos/virologia , Receptores de Reconhecimento de Padrão/genética , Receptores de Reconhecimento de Padrão/imunologia , Linfócitos T/imunologia , Linfócitos T/virologia , Vírion/genética , Viroses/virologia , Vírus/genética
11.
Virology ; 492: 130-9, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26922001

RESUMO

Autophagy is a cellular process used to eliminate intracellular pathogens. Many viruses however are able to manipulate this cellular process for their own advantage. Here we demonstrate that Mouse Norovirus (MNV) infection induces autophagy but does not appear to utilise the autophagosomal membrane for establishment and formation of the viral replication complex. We have observed that MNV infection results in lipidation and recruitment of LC3 to the autophagosome membrane but prevents subsequent fusion of the autophagosomes with lysosomes, as SQSTM1 (an autophagy receptor) accumulates and Lysosome-Associated Membrane Protein1 is sequestered to the MNV replication complex (RC) rather than to autophagosomes. We have additionally observed that chemical modulation of autophagy differentially affects MNV replication. From this study we can conclude that MNV infection induces autophagy, however suppresses the final maturation step of this response, indicating that autophagy induction contributes to MNV replication independently of RC biogenesis.


Assuntos
Autofagia/genética , Interações Hospedeiro-Patógeno , Macrófagos/virologia , Proteínas Associadas aos Microtúbulos/genética , Norovirus/genética , Fagossomos/virologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Regulação da Expressão Gênica , Células HEK293 , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/ultraestrutura , Membranas Intracelulares/virologia , Proteínas de Membrana Lisossomal/genética , Proteínas de Membrana Lisossomal/metabolismo , Macrófagos/citologia , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Norovirus/metabolismo , Fagossomos/metabolismo , Fagossomos/ultraestrutura , Proteína Sequestossoma-1 , Transdução de Sinais , Células Vero , Replicação Viral/genética
12.
Cell Microbiol ; 18(3): 355-68, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26332641

RESUMO

Environmental mycobacteria, highly prevalent in natural and artificial (including chlorinated municipal water) niches, are emerging as new threat to human health, especially to HIV-infected population. These seemingly harmless non-pathogenic mycobacteria, which are otherwise cleared, establish as opportunistic infections adding to HIV-associated complications. Although immune-evading strategies of pathogenic mycobacteria are known, the mechanisms underlying the early events by which opportunistic mycobacteria establish infection in macrophages and influencing HIV infection are unclear. Proteomics of phagosome-enriched fractions from Mycobacterium bovis Bacillus Calmette-Guérin (BCG) mono-infected and HIV-M. bovis BCG co-infected THP-1 cells by LC-MALDI-MS/MS revealed differential distribution of 260 proteins. Validation of the proteomics data showed that HIV co-infection helped the survival of non-pathogenic mycobacteria by obstructing phagosome maturation, promoting lipid biogenesis and increasing intracellular ATP equivalents. In turn, mycobacterial co-infection up-regulated purinergic receptors in macrophages that are known to support HIV entry, explaining increased viral titers during co-infection. The mutualism was reconfirmed using clinically relevant opportunistic mycobacteria, Mycobacterium avium, Mycobacterium kansasii and Mycobacterium phlei that exhibited increased survival during co-infection, together with increase in HIV titers. Additionally, the catalogued proteins in the study provide new leads that will significantly add to the understanding of the biology of opportunistic mycobacteria and HIV coalition.


Assuntos
Coinfecção/microbiologia , Coinfecção/virologia , Infecções por HIV/microbiologia , Infecções por Mycobacterium/virologia , Trifosfato de Adenosina/metabolismo , Linhagem Celular , Coinfecção/metabolismo , Citocinas/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Macrófagos/microbiologia , Macrófagos/virologia , Mycobacterium/patogenicidade , Mycobacterium bovis/patogenicidade , Fagossomos/microbiologia , Fagossomos/virologia , Proteômica/métodos , Simbiose , Carga Viral
13.
Biosci Trends ; 9(2): 111-6, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25971696

RESUMO

In this study, we investigate the relationship of hepatitis B virus (HBV) infection and autophagy. HepG2 cells and HepG2 cells infected with HBV (HepG2.2.15) were transfected with GFP-LC3 (green fluorescence protein conjugated with microtubule-associated protein 1 light chain 3) expression vector and autophagy status was then examined with confocal microscope. HepG2.2.15 cells were further treated with serum-free medium or 3-methyladenine (3-MA), and subjected to Hepatitis B core antigen (HBcAg), Hepatitis B surface antigen (HBsAg), or hepatitis B polymerase protein detection by immunohistochemistry. Localization of the GFP-LC3 and the HBV proteins was observed by confocal fluorescence microscope. The level of SQSTM1/p62 protein was also evaluated by Western blot analysis. In contrast to a diffuse distribution in HepG2 cells, GFP-LC3 formed distinct punctate dots, which were further enhanced by nutritional starvation, in HepG2.2.15 cells. The expression of hepatitis B polymerase and HBcAg, but not HBsAg, was positively correlated with the autophagic intensity. However, no co-localizations were observed between HBV proteins and autophagosomes. Suppression of autophagy reduced the expression of hepatitis B polymerase and HBcAg, but not HBsAg. Western blot showed that SQSTM1/p62 protein level was declined in HepG2.2.15 cells comparing HepG2 cells, and further reduced while upon serum starvation. In conclusion, HBV infection induces autophagic degradation and autophagy. Autophagy is critical for HBV replication. However HBV replication does not take place in autophagosomes.


Assuntos
Autofagia , Vírus da Hepatite B/fisiologia , Fagossomos/virologia , Replicação Viral/fisiologia , Células Hep G2 , Antígenos de Superfície da Hepatite B/metabolismo , Humanos , Transporte Proteico , Vacúolos/metabolismo , Proteínas Virais/metabolismo
14.
Traffic ; 16(8): 814-31, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25869659

RESUMO

The prototypic poxvirus, vaccinia virus (VACV), occurs in two infectious forms, mature virions (MVs) and extracellular virions (EVs). Both enter HeLa cells by inducing macropinocytic uptake. Using confocal microscopy, live-cell imaging, targeted RNAi screening and perturbants of endosome maturation, we analyzed the properties and maturation pathway of the macropinocytic vacuoles containing VACV MVs in HeLa cells. The vacuoles first acquired markers of early endosomes [Rab5, early endosome antigen 1 and phosphatidylinositol(3)P]. Prior to release of virus cores into the cytoplasm, they contained markers of late endosomes and lysosomes (Rab7a, lysosome-associated membrane protein 1 and sorting nexin 3). RNAi screening of endocytic cell factors emphasized the importance of late compartments for VACV infection. Follow-up perturbation analysis showed that infection required Rab7a and PIKfyve, confirming that VACV is a late-penetrating virus dependent on macropinosome maturation. VACV EV infection was inhibited by depletion of many of the same factors, indicating that both infectious particle forms share the need for late vacuolar conditions for penetration.


Assuntos
Fagocitose , Fagossomos/metabolismo , Vaccinia virus/patogenicidade , Endossomos/metabolismo , Endossomos/virologia , Células HeLa , Humanos , Proteína 1 de Membrana Associada ao Lisossomo/genética , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Fagossomos/virologia , Nexinas de Classificação/genética , Nexinas de Classificação/metabolismo , Vaccinia virus/metabolismo , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab5 de Ligação ao GTP/genética , Proteínas rab5 de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
15.
Oncogene ; 34(3): 334-45, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24362528

RESUMO

The retroviral oncoprotein Tax from human T-cell leukemia virus type 1 (HTLV-1), an etiological factor that causes adult T-cell leukemia and lymphoma, has a crucial role in initiating T-lymphocyte transformation by inducing oncogenic signaling activation. We here report that Tax is a determining factor for dysregulation of autophagy in HTLV-1-transformed T cells and Tax-immortalized CD4 memory T cells. Tax facilitated autophagic process by activating inhibitor of κB (IκB) kinase (IKK) complex, which subsequently recruited an autophagy molecular complex containing Beclin1 and Bif-1 to the lipid raft microdomains. Tax engaged a crosstalk between IKK complex and autophagic molecule complex by directly interacting with both complexes, promoting assembly of LC3+ autophagosomes. Moreover, expression of lipid raft-targeted Bif-1 or Beclin1 was sufficient to induce formation of LC3+ autophagosomes, suggesting that Tax recruitment of autophagic molecules to lipid rafts is a dominant strategy to deregulate autophagy in the context of HTLV-1 transformation of T cells. Furthermore, depletion of autophagy molecules such as Beclin1 and PI3 kinase class III resulted in impaired growth of HTLV-1-transformed T cells, indicating a critical role of Tax-deregulated autophagy in promoting survival and transformation of virally infected T cells.


Assuntos
Autofagia , Produtos do Gene tax/metabolismo , Vírus Linfotrópico T Tipo 1 Humano/metabolismo , Microdomínios da Membrana/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Linhagem Celular Tumoral , Células Cultivadas , Produtos do Gene tax/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Interações Hospedeiro-Patógeno , Vírus Linfotrópico T Tipo 1 Humano/genética , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Humanos , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Immunoblotting , Células Jurkat , Microdomínios da Membrana/virologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Microscopia de Fluorescência , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação , Fagossomos/metabolismo , Fagossomos/virologia , Fosfatidilinositol 3-Quinases , Linfócitos T/metabolismo , Linfócitos T/virologia
16.
Protein Cell ; 5(12): 912-27, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25311841

RESUMO

Autophagy plays important roles in modulating viral replication and antiviral immune response. Coronavirus infection is associated with the autophagic process, however, little is known about the mechanisms of autophagy induction and its contribution to coronavirus regulation of host innate responses. Here, we show that the membrane-associated papain-like protease PLP2 (PLP2-TM) of coronaviruses acts as a novel autophagy-inducing protein. Intriguingly, PLP2-TM induces incomplete autophagy process by increasing the accumulation of autophagosomes but blocking the fusion of autophagosomes with lysosomes. Furthermore, PLP2-TM interacts with the key autophagy regulators, LC3 and Beclin1, and promotes Beclin1 interaction with STING, the key regulator for antiviral IFN signaling. Finally, knockdown of Beclin1 partially reverses PLP2-TM's inhibitory effect on innate immunity which resulting in decreased coronavirus replication. These results suggested that coronavirus papain-like protease induces incomplete autophagy by interacting with Beclin1, which in turn modulates coronavirus replication and antiviral innate immunity.


Assuntos
Proteínas Reguladoras de Apoptose/imunologia , Coronavirus Humano NL63/imunologia , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/imunologia , Proteínas de Membrana/imunologia , Proteínas Associadas aos Microtúbulos/imunologia , Papaína/imunologia , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Proteínas Reguladoras de Apoptose/genética , Autofagia , Proteína Beclina-1 , Coronavirus Humano NL63/genética , Proteases Semelhantes à Papaína de Coronavírus , Células HEK293 , Células HeLa , Humanos , Evasão da Resposta Imune , Imunidade Inata , Interferon gama/genética , Interferon gama/imunologia , Lisossomos/metabolismo , Lisossomos/virologia , Células MCF-7 , Fusão de Membrana , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Proteínas Associadas aos Microtúbulos/genética , Papaína/genética , Fagossomos/metabolismo , Fagossomos/virologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/imunologia , Transdução de Sinais , Replicação Viral
17.
Infect Genet Evol ; 26: 95-102, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24836289

RESUMO

Coxsackievirus B3 (CVB3) is known to induce both autophagy and apoptosis, but whether a relationship exists between these processes upon infection, and whether and how they influence the viral life cycle are currently unknown. We observed here that while autophagosome formation increased in CVB3-infected HeLa cells at the early stage of infection, it decreased at the late stage of infection along with increased apoptosis. Examining whether a functional relationship existed between autophagy and apoptosis during CVB3 infection, we found that increasing levels of autophagy inhibited apoptosis and that some apoptotic proteins in the endogenous and exogenous apoptosis pathways played a role in the transition from autophagy to apoptosis by cleaving the autophagy-related proteins Beclin-1 and Atg5. However, the transcription and translation of full-length Atg5 and Beclin-1 also increased, which likely counteracted the cleavage effect in order to prevent cells from dying too early and to ensure that CVB3 replication was complete in the autophagosomes. Using pharmacological inducers and inhibitors of autophagy as well as inhibitors of apoptosis, we found that while CVB3 replication relied on the autophagosomes, its release from the cell depended on apoptosis. Therefore, autophagy and apoptosis are two important processes that interact with each other during CVB3 infection, promoting the CVB3 life cycle.


Assuntos
Apoptose , Autofagia , Caspases/metabolismo , Enterovirus Humano B/fisiologia , Fagossomos/metabolismo , Fagossomos/virologia , Liberação de Vírus , Replicação Viral , Linhagem Celular , Infecções por Coxsackievirus/genética , Infecções por Coxsackievirus/metabolismo , Infecções por Coxsackievirus/virologia , Humanos , Transcrição Gênica
18.
PLoS Pathog ; 10(4): e1004045, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24722773

RESUMO

Coxsackievirus B3 (CVB3), a member of the picornavirus family and enterovirus genus, causes viral myocarditis, aseptic meningitis, and pancreatitis in humans. We genetically engineered a unique molecular marker, "fluorescent timer" protein, within our infectious CVB3 clone and isolated a high-titer recombinant viral stock (Timer-CVB3) following transfection in HeLa cells. "Fluorescent timer" protein undergoes slow conversion of fluorescence from green to red over time, and Timer-CVB3 can be utilized to track virus infection and dissemination in real time. Upon infection with Timer-CVB3, HeLa cells, neural progenitor and stem cells (NPSCs), and C2C12 myoblast cells slowly changed fluorescence from green to red over 72 hours as determined by fluorescence microscopy or flow cytometric analysis. The conversion of "fluorescent timer" protein in HeLa cells infected with Timer-CVB3 could be interrupted by fixation, suggesting that the fluorophore was stabilized by formaldehyde cross-linking reactions. Induction of a type I interferon response or ribavirin treatment reduced the progression of cell-to-cell virus spread in HeLa cells or NPSCs infected with Timer-CVB3. Time lapse photography of partially differentiated NPSCs infected with Timer-CVB3 revealed substantial intracellular membrane remodeling and the assembly of discrete virus replication organelles which changed fluorescence color in an asynchronous fashion within the cell. "Fluorescent timer" protein colocalized closely with viral 3A protein within virus replication organelles. Intriguingly, infection of partially differentiated NPSCs or C2C12 myoblast cells induced the release of abundant extracellular microvesicles (EMVs) containing matured "fluorescent timer" protein and infectious virus representing a novel route of virus dissemination. CVB3 virions were readily observed within purified EMVs by transmission electron microscopy, and infectious virus was identified within low-density isopycnic iodixanol gradient fractions consistent with membrane association. The preferential detection of the lipidated form of LC3 protein (LC3 II) in released EMVs harboring infectious virus suggests that the autophagy pathway plays a crucial role in microvesicle shedding and virus release, similar to a process previously described as autophagosome-mediated exit without lysis (AWOL) observed during poliovirus replication. Through the use of this novel recombinant virus which provides more dynamic information from static fluorescent images, we hope to gain a better understanding of CVB3 tropism, intracellular membrane reorganization, and virus-associated microvesicle dissemination within the host.


Assuntos
Micropartículas Derivadas de Células/virologia , Enterovirus Humano B/fisiologia , Infecções por Enterovirus/metabolismo , Fagossomos/virologia , Eliminação de Partículas Virais/fisiologia , Animais , Micropartículas Derivadas de Células/genética , Micropartículas Derivadas de Células/metabolismo , Infecções por Enterovirus/genética , Células HeLa , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Fagossomos/genética , Fagossomos/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
19.
PLoS Pathog ; 9(12): e1003722, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24339771

RESUMO

Human hepatitis B virus (HBV) causes chronic hepatitis and is associated with the development of hepatocellular carcinoma. HBV infection alters mitochondrial metabolism. The selective removal of damaged mitochondria is essential for the maintenance of mitochondrial and cellular homeostasis. Here, we report that HBV shifts the balance of mitochondrial dynamics toward fission and mitophagy to attenuate the virus-induced apoptosis. HBV induced perinuclear clustering of mitochondria and triggered mitochondrial translocation of the dynamin-related protein (Drp1) by stimulating its phosphorylation at Ser616, leading to mitochondrial fission. HBV also stimulated the gene expression of Parkin, PINK1, and LC3B and induced Parkin recruitment to the mitochondria. Upon translocation to mitochondria, Parkin, an E3 ubiquitin ligase, underwent self-ubiquitination and facilitated the ubiquitination and degradation of its substrate Mitofusin 2 (Mfn2), a mediator of mitochondrial fusion. In addition to conventional immunofluorescence, a sensitive dual fluorescence reporter expressing mito-mRFP-EGFP fused in-frame to a mitochondrial targeting sequence was employed to observe the completion of the mitophagic process by delivery of the engulfed mitochondria to lysosomes for degradation. Furthermore, we demonstrate that viral HBx protein plays a central role in promoting aberrant mitochondrial dynamics either when expressed alone or in the context of viral genome. Perturbing mitophagy by silencing Parkin led to enhanced apoptotic signaling, suggesting that HBV-induced mitochondrial fission and mitophagy promote cell survival and possibly viral persistence. Altered mitochondrial dynamics associated with HBV infection may contribute to mitochondrial injury and liver disease pathogenesis.


Assuntos
Apoptose , Vírus da Hepatite B/fisiologia , Hepatite B/virologia , Dinâmica Mitocondrial , Mitofagia , Apoptose/genética , Células Cultivadas , Dinaminas , GTP Fosfo-Hidrolases/metabolismo , Regulação da Expressão Gênica , Células Hep G2 , Hepatite B/genética , Hepatite B/patologia , Vírus da Hepatite B/patogenicidade , Humanos , Proteínas Associadas aos Microtúbulos/metabolismo , Dinâmica Mitocondrial/genética , Proteínas Mitocondriais/metabolismo , Mitofagia/genética , Fagossomos/virologia , Transporte Proteico , Transativadores/fisiologia , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Virais Reguladoras e Acessórias
20.
Essays Biochem ; 55: 153-63, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24070478

RESUMO

Autophagy is a membrane trafficking pathway that results in the formation of autophagosomes which deliver portions of the cytosol to lysosomes for degradation. When autophagosomes engulf intracellular pathogens, the pathway is called 'xenophagy' because it leads to the removal of foreign material. Autophagy is activated during infection by Toll-like receptors that recognize pathogen-associated molecular patterns. This allows autophagy to kill micro-organisms and present pathogen components to the innate and acquired immune systems. The targeting of pathogens by autophagy is selective and involves a growing family of autophagy receptors that bind to the autophagosome membrane protein LC3 (light-chain 3)/Atg8 (autography-related protein 8). Ubiquitination of microbes identifies them as substrates for autophagy and they are delivered to autophagosomes by autophagy receptors that bind both ubiquitin and LC3/Atg8. Bacteria can also be detected before they enter the cytosol by autophagy receptors that scan the surface of membrane compartments for evidence of damage. The observation that some pathogens survive in cells suggests they can evade complete destruction by autophagy. For some bacteria this involves proteins that shield the surface of the bacteria from recognition by autophagy receptors. Other viruses and bacteria are resistant to degradation in lysosomes and use autophagosomes and/or lysosomes as sites for replication. Most of our current understanding of the role played by autophagy during microbial infection has come from studies of bacteria and viruses in tissue culture cell lines. Future work will focus on understanding how autophagy determines the outcome of infection 'in vivo', and how autophagy pathways can be exploited therapeutically.


Assuntos
Autofagia , Interações Hospedeiro-Patógeno , Fagossomos/metabolismo , Animais , Bactérias/patogenicidade , Humanos , Lisossomos/metabolismo , Lisossomos/microbiologia , Lisossomos/virologia , Fagossomos/microbiologia , Fagossomos/virologia , Vírus/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA