Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 98(8): e0032724, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39082785

RESUMEN

African swine fever (ASF), caused by the African swine fever virus (ASFV), is a highly infectious disease afflicting domestic pigs and wild boars. It exhibits an alarming acute infection fatality rate of up to 100%. Regrettably, no commercial vaccines or specific drugs for combating this disease are currently available. This study evaluated the anti-ASFV activities in porcine alveolar macrophages, 3D4/21 cells, and PK-15 cells of four bis-benzylisoquinoline alkaloids (BBAs): cepharanthine (CEP), tetrandrine, fangchinoline, and iso-tetrandrine. Furthermore, we demonstrated that CEP, which exhibited the highest selectivity index (SI = 81.31), alkalized late endosomes/lysosomes, hindered ASFV endosomal transport, disrupted virus uncoating signals, and thereby inhibited ASFV internalization. Additionally, CEP disrupted ASFV DNA synthesis, leading to the inhibition of viral replication. Moreover, berbamine was labeled with NBD to synthesize a fluorescent probe to study the cellular location of these BBAs. By co-staining with Lyso-Tracker and lysosome-associated membrane protein 1, we demonstrated that BBAs target the endolysosomal compartments for the first time. Our data together indicated that BBAs are a class of natural products with significant inhibitory effects against ASFV infection. These findings suggest their potential efficacy as agents for the prevention and control of ASF, offering valuable references for the identification of potential drug targets.IMPORTANCEThe urgency and severity of African swine fever (ASF) underscore the critical need for effective interventions against this highly infectious disease, which poses a grave threat to domestic pigs and wild boars. Our study reveals the potent anti-African swine fever virus (ASFV) efficacy of bis-benzylisoquinoline alkaloids (BBAs), particularly evident in the absence of progeny virus production under a 5 µM concentration treatment. The structural similarity among cepharanthine, tetrandrine, fangchinoline, and iso-tetrandrine, coupled with their analogous inhibitory stages and comparable selectivity indexes, strongly suggests a shared antiviral mechanism within this drug category. Further investigation revealed that BBAs localize to lysosomes and inhibit the internalization and replication of ASFV by disrupting the endosomal/lysosomal function. These collective results have profound implications for ASF prevention and control, suggesting the potential of the investigated agents as prophylactic and therapeutic measures. Furthermore, our study offers crucial insights into identifying drug targets and laying the groundwork for innovative interventions.


Asunto(s)
Virus de la Fiebre Porcina Africana , Antivirales , Bencilisoquinolinas , Endosomas , Lisosomas , Internalización del Virus , Replicación Viral , Animales , Virus de la Fiebre Porcina Africana/efectos de los fármacos , Virus de la Fiebre Porcina Africana/fisiología , Internalización del Virus/efectos de los fármacos , Bencilisoquinolinas/farmacología , Replicación Viral/efectos de los fármacos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Lisosomas/virología , Porcinos , Endosomas/metabolismo , Endosomas/efectos de los fármacos , Endosomas/virología , Antivirales/farmacología , Línea Celular , Fiebre Porcina Africana/virología , Fiebre Porcina Africana/tratamiento farmacológico , Fiebre Porcina Africana/metabolismo , Guanina/análogos & derivados , Guanina/farmacología , Alcaloides/farmacología , Macrófagos Alveolares/virología , Macrófagos Alveolares/efectos de los fármacos , Benzodioxoles
2.
J Virol ; 97(6): e0054923, 2023 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-37222617

RESUMEN

Porcine epidemic diarrhea virus (PEDV) has caused huge economic losses to the global pig industry. The swine enteric coronavirus spike (S) protein recognizes various cell surface molecules to regulate viral infection. In this study, we identified 211 host membrane proteins related to the S1 protein by pulldown combined with liquid-chromatography tandem mass spectrometry (LC-MS/MS) analysis. Among these, heat shock protein family A member 5 (HSPA5) was identified through screening as having a specific interaction with the PEDV S protein, and positive regulation of PEDV infection was validated by knockdown and overexpression tests. Further studies verified the role of HSPA5 in viral attachment and internalization. In addition, we found that HSPA5 interacts with S proteins through its nucleotide-binding structural domain (NBD) and that polyclonal antibodies can block viral infection. In detail, HSPA5 was found to be involved in viral trafficking via the endo-/lysosomal pathway. Inhibition of HSPA5 activity during internalization would reduce the subcellular colocalization of PEDV with lysosomes in the endo-/lysosomal pathway. Together, these findings show that HSPA5 is a novel PEDV potential target for the creation of therapeutic drugs. IMPORTANCE PEDV infection causes severe piglet mortality and threatens the global pig industry. However, the complex invasion mechanism of PEDV makes its prevention and control difficult. Here, we determined that HSPA5 is a novel target for PEDV which interacts with its S protein and is involved in viral attachment and internalization, influencing its transport via the endo-/lysosomal pathway. Our work extends knowledge about the relationship between the PEDV S and host proteins and provides a new therapeutic target against PEDV infection.


Asunto(s)
Infecciones por Coronavirus , Chaperón BiP del Retículo Endoplásmico , Virus de la Diarrea Epidémica Porcina , Glicoproteína de la Espiga del Coronavirus , Enfermedades de los Porcinos , Internalización del Virus , Animales , Chlorocebus aethiops , Infecciones por Coronavirus/fisiopatología , Infecciones por Coronavirus/virología , Lisosomas/metabolismo , Lisosomas/virología , Virus de la Diarrea Epidémica Porcina/genética , Virus de la Diarrea Epidémica Porcina/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Porcinos , Enfermedades de los Porcinos/fisiopatología , Enfermedades de los Porcinos/virología , Células Vero , Chaperón BiP del Retículo Endoplásmico/genética , Chaperón BiP del Retículo Endoplásmico/metabolismo , Acoplamiento Viral , Endocitosis/genética
3.
J Virol ; 97(12): e0133823, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-38009916

RESUMEN

IMPORTANCE: Betacoronaviruses, including severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and mouse hepatitis virus (MHV), exploit the lysosomal exocytosis pathway for egress. However, whether all betacoronaviruses members use the same pathway to exit cells remains unknown. Here, we demonstrated that porcine hemagglutinating encephalomyelitis virus (PHEV) egress occurs by Arl8b-dependent lysosomal exocytosis, a cellular egress mechanism shared by SARS-CoV-2 and MHV. Notably, PHEV acidifies lysosomes and activates lysosomal degradative enzymes, while SARS-CoV-2 and MHV deacidify lysosomes and limit the activation of lysosomal degradative enzymes. In addition, PHEV release depends on V-ATPase-mediated lysosomal pH. Furthermore, this is the first study to evaluate ßCoV using lysosome for spreading through the body, and we have found that lysosome played a critical role in PHEV neural transmission and brain damage caused by virus infection in the central nervous system. Taken together, different betacoronaviruses could disrupt lysosomal function differently to exit cells.


Asunto(s)
Betacoronavirus 1 , Infecciones por Coronavirus , Exocitosis , Lisosomas , Neuronas , Animales , Ratones , Betacoronavirus 1/metabolismo , Lisosomas/enzimología , Lisosomas/metabolismo , Lisosomas/virología , Virus de la Hepatitis Murina/metabolismo , Neuronas/enzimología , Neuronas/metabolismo , Neuronas/patología , Neuronas/virología , SARS-CoV-2/metabolismo , Porcinos/virología , Concentración de Iones de Hidrógeno , ATPasas de Translocación de Protón Vacuolares/metabolismo , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/transmisión , Infecciones por Coronavirus/virología
4.
J Med Virol ; 96(9): e29906, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39262090

RESUMEN

Influenza virus-induced viral pneumonia is a major threat to human health, and specific therapeutic agents for viral pneumonia are still lacking. MoringaA (MA) is an anti-influenza virus active compound isolated from Moringa seeds, which can inhibit influenza virus by activating the TFEB-autophagic lysosomal pathway in host cells. In this study, we obtained exosomes from M2-type macrophages and encapsulated and delivered MA (MA-Exos), and we investigated the efficacy of MA-Exos in antiviral and viral pneumonia in vivo and in vitro, respectively. In addition, we provided insights into the mechanism by which MA-Exos regulates TFEB-lysosomal autophagy by RNA sequencing. The MA-Exos showed broad-spectrum inhibition of IAV, and significant promotion of the autophagic lysosomal pathway. Meanwhile, we found that GCN5 gene and protein were significantly down-regulated in IAV-infected cells after MA-Exos intervention, indicating its blocking the acetylation of TFEB by GCN5. In addition, MA-Exos also significantly promoted autophagy in lung tissue cells of mice with viral pneumonia. MA-Exos can inhibit and clear influenza virus by mediating the TFEB-autophagy lysosomal pathway by a mechanism related to the down-regulation of histone acetyltransferase GCN5. Our study provides a strategy for targeting MA-Exos for the treatment of viral pneumonia from both antiviral and virus-induced inflammation inhibition pathways.


Asunto(s)
Antivirales , Autofagia , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Exosomas , Virus de la Influenza A , Lisosomas , Animales , Ratones , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Lisosomas/metabolismo , Lisosomas/efectos de los fármacos , Lisosomas/virología , Exosomas/metabolismo , Antivirales/farmacología , Autofagia/efectos de los fármacos , Humanos , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/fisiología , Infecciones por Orthomyxoviridae/virología , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Macrófagos/virología , Macrófagos/efectos de los fármacos , Pulmón/virología
5.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34021074

RESUMEN

COVID-19, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a global pandemic and has claimed over 2 million lives worldwide. Although the genetic sequences of SARS-CoV and SARS-CoV-2 have high homology, the clinical and pathological characteristics of COVID-19 differ significantly from those of SARS. How and whether SARS-CoV-2 evades (cellular) immune surveillance requires further elucidation. In this study, we show that SARS-CoV-2 infection leads to major histocompability complex class Ι (MHC-Ι) down-regulation both in vitro and in vivo. The viral protein encoded by open reading frame 8 (ORF8) of SARS-CoV-2, which shares the least homology with SARS-CoV among all viral proteins, directly interacts with MHC-Ι molecules and mediates their down-regulation. In ORF8-expressing cells, MHC-Ι molecules are selectively targeted for lysosomal degradation via autophagy. Thus, SARS-CoV-2-infected cells are much less sensitive to lysis by cytotoxic T lymphocytes. Because ORF8 protein impairs the antigen presentation system, inhibition of ORF8 could be a strategy to improve immune surveillance.


Asunto(s)
Presentación de Antígeno , COVID-19/inmunología , Regulación hacia Abajo/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Evasión Inmune , SARS-CoV-2/inmunología , Proteínas Virales/inmunología , Animales , Autofagia/genética , Autofagia/inmunología , COVID-19/genética , Chlorocebus aethiops , Células HEK293 , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Lisosomas/genética , Lisosomas/inmunología , Lisosomas/virología , Ratones , Ratones Transgénicos , SARS-CoV-2/genética , Células Vero , Proteínas Virales/genética
6.
J Virol ; 96(14): e0076722, 2022 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-35770989

RESUMEN

Production of infectious HIV-1 particles requires incorporation of the viral envelope glycoprotein (Env) at the plasma membrane (PM) of infected CD4+ T cells. Env trafficking to the PM exposes viral epitopes that can be exploited by the host immune system; however, HIV-1 can evade this response by endocytosis of excess Env from the PM. The fate of Env after internalization remains unclear, with evidence suggesting several different vesicular trafficking steps may be involved, including recycling pathways. To date, there have been very few studies documenting the trafficking pathways of native Env in infected T cells. Furthermore, it remains unclear whether there are T-cell-specific endosomal pathways regulating the fate of endocytic Env. Here, we use a pulse-labeling approach with a monovalent anti-Env Fab probe to characterize the trafficking of internalized Env within infected CD4+ T-cell lines, together with CRISPR/Cas9-mediated endogenous protein tagging, to assess the role of host cell Rab GTPases in Env trafficking. We show that endocytosed Env traffics to Rab14+ compartments that possess hallmarks of late endosomes and lysosomes. We also demonstrate that Env can recycle back to the PM, although we find that recycling does not occur at high rates when compared to the model recycling protein transferrin. These results help to resolve open questions about the fate and relevance of endocytosed Env in HIV-infected cells and suggest a novel role for Rab14 in a cell-type-specific late-endosomal/lysosomal trafficking pathway in T cells. IMPORTANCE HIV-1 envelope glycoprotein (Env) evades immune neutralization through many mechanisms. One immune evasion strategy may result from the internalization of excess surface-exposed Env to prevent antibody-dependent cellular cytotoxicity or neutralization. Characterization of the fate of endocytosed Env is critical to understand which vesicular pathways could be targeted to promote display of Env epitopes to the immune system. In this study, we characterize the endocytic fate of native Env, expressed from infected human T-cell lines. We demonstrate that Env is rapidly trafficked to a late-endosome/lysosome-like compartment and can be recycled to the cell surface for incorporation into virus assembly sites. This study implicates a novel intracellular compartment, marked by host-cell Rab14 GTPases, for the sequestration of Env. Therapeutic approaches aimed at mobilizing this intracellular pool of Env could lead to stronger immune control of HIV-1 infection via antibody-dependent cell-mediated cytotoxicity.


Asunto(s)
Endosomas , Infecciones por VIH , VIH-1 , Lisosomas , Linfocitos T , Productos del Gen env del Virus de la Inmunodeficiencia Humana , Línea Celular , Endocitosis , Endosomas/metabolismo , Endosomas/virología , Epítopos , Infecciones por VIH/metabolismo , Humanos , Lisosomas/metabolismo , Lisosomas/virología , Transporte de Proteínas , Linfocitos T/virología , Productos del Gen env del Virus de la Inmunodeficiencia Humana/metabolismo , Proteínas de Unión al GTP rab/metabolismo
7.
PLoS Pathog ; 17(2): e1009312, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33539432

RESUMEN

Many small molecules have been identified as entry inhibitors of filoviruses. However, a lack of understanding of the mechanism of action for these molecules limits further their development as anti-filoviral agents. Here we provide evidence that toremifene and other small molecule entry inhibitors have at least three distinctive mechanisms of action and lay the groundwork for future development of anti-filoviral agents. The three mechanisms identified here include: (1) direct binding to the internal fusion loop region of Ebola virus glycoprotein (GP); (2) the HR2 domain is likely the main binding site for Marburg virus GP inhibitors and a secondary binding site for some EBOV GP inhibitors; (3) lysosome trapping of GP inhibitors increases drug exposure in the lysosome and further improves the viral inhibition. Importantly, small molecules targeting different domains on GP are synergistic in inhibiting EBOV entry suggesting these two mechanisms of action are distinct. Our findings provide important mechanistic insights into filovirus entry and rational drug design for future antiviral development.


Asunto(s)
Antivirales/farmacología , Ebolavirus/efectos de los fármacos , Glicoproteínas/metabolismo , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/farmacología , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus/efectos de los fármacos , Células A549 , Animales , Chlorocebus aethiops , Ebolavirus/fisiología , Glicoproteínas/genética , Fiebre Hemorrágica Ebola/metabolismo , Fiebre Hemorrágica Ebola/patología , Fiebre Hemorrágica Ebola/virología , Interacciones Huésped-Patógeno , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/virología , Células Vero , Proteínas del Envoltorio Viral/genética
8.
J Immunol ; 206(6): 1284-1296, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33568400

RESUMEN

Neutralizing Abs suppress HIV infection by accelerating viral clearance from blood circulation in addition to neutralization. The elimination mechanism is largely unknown. We determined that human liver sinusoidal endothelial cells (LSEC) express FcγRIIb as the lone Fcγ receptor, and using humanized FcγRIIb mouse, we found that Ab-opsonized HIV pseudoviruses were cleared considerably faster from circulation than HIV by LSEC FcγRIIb. Compared with humanized FcγRIIb-expressing mice, HIV clearance was significantly slower in FcγRIIb knockout mice. Interestingly, a pentamix of neutralizing Abs cleared HIV faster compared with hyperimmune anti-HIV Ig (HIVIG), although the HIV Ab/Ag ratio was higher in immune complexes made of HIVIG and HIV than pentamix and HIV. The effector mechanism of LSEC FcγRIIb was identified to be endocytosis. Once endocytosed, both Ab-opsonized HIV pseudoviruses and HIV localized to lysosomes. This suggests that clearance of HIV, endocytosis, and lysosomal trafficking within LSEC occur sequentially and that the clearance rate may influence downstream events. Most importantly, we have identified LSEC FcγRIIb-mediated endocytosis to be the Fc effector mechanism to eliminate cell-free HIV by Abs, which could inform development of HIV vaccine and Ab therapy.


Asunto(s)
Anticuerpos Neutralizantes/metabolismo , Endocitosis/inmunología , Células Endoteliales/inmunología , Infecciones por VIH/inmunología , Receptores de IgG/metabolismo , Animales , Capilares/citología , Capilares/inmunología , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/virología , Endotelio Vascular/citología , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Células HEK293 , VIH/inmunología , Infecciones por VIH/sangre , Infecciones por VIH/patología , Infecciones por VIH/virología , Voluntarios Sanos , Humanos , Hígado/irrigación sanguínea , Hígado/inmunología , Lisosomas/metabolismo , Lisosomas/virología , Masculino , Ratones , Ratones Noqueados , Cultivo Primario de Células , Receptores de IgG/genética
9.
J Virol ; 95(6)2021 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-33328308

RESUMEN

Classical swine fever (CSF), caused by classical swine fever virus (CSFV), is a highly contagious disease of swine with high morbidity and mortality that negatively affects the pig industry worldwide, in particular in China. Soon after the endocytosis of CSFV, the virus makes full use of the components of host cells to complete its life cycle. The endocytosis sorting complex required for transport (ESCRT) system is a central molecular machine for membrane protein sorting and scission in eukaryotic cells that plays an essential role in many physiological metabolic processes, including invasion and egress of envelope viruses. However, the molecular mechanism that ESCRT uses to regulate the replication of CSFV is unknown. In this study, we demonstrated that the ESCRT-I complex Tsg101 protein participates in clathrin-mediated endocytosis of CSFV and is also involved in CSFV trafficking. Tsg101 assists the virus in entering the host cell through the late endosome (Rab7 and Rab9) and finally reaching the lysosome (Lamp-1). Interestingly, Tsg101 is also involved in the viral replication process by interacting with nonstructural proteins 4B and 5B of CSFV. Finally, confocal microscopy showed that the replication complex of Tsg101 and double-stranded RNA (dsRNA) or NS4B and NS5B protein was close to the endoplasmic reticulum (ER), not the Golgi, in the cytoplasm. Collectively, our finding highlights that Tsg101 regulates the process of CSFV entry and replication, indicating that the ESCRT plays an important role in the life cycle of CSFV. Thus, ESCRT molecules could serve as therapeutic targets to combat CSFV infection.IMPORTANCE CSF, caused by CSFV, is a World Organization for Animal Health (OIE) notifiable disease and causes significant financial losses to the pig industry globally. The ESCRT machinery plays an important regulatory role in several members of the genera Flavivirus and Hepacivirus within the family Flaviviridae, such as hepatitis C virus, Japanese encephalitis virus, and dengue virus. Previous reports have shown that assembling and budding of these viruses require ESCRT. However, the role of ESCRT in Pestivirus infection remains to be elucidated. We determined the molecular mechanisms of the regulation of CSFV infection by the major subunit Tsg101 of ESCRT-I. Interestingly, Tsg101 plays an essential regulatory role in both clathrin-mediated endocytosis and genome replication of CSFV. Overall, the results of this study provide further insights into the molecular function of ESCRT-I complex protein Tsg101 during CSFV infection, which may serve as a molecular target for pestivirus inhibitors.


Asunto(s)
Virus de la Fiebre Porcina Clásica/fisiología , Proteínas de Unión al ADN/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Factores de Transcripción/metabolismo , Internalización del Virus , Replicación Viral , Animales , Línea Celular , Peste Porcina Clásica/metabolismo , Peste Porcina Clásica/virología , Proteínas de Unión al ADN/genética , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Endosomas/metabolismo , Endosomas/virología , Interacciones Huésped-Patógeno , Lisosomas/metabolismo , Lisosomas/virología , ARN Viral/metabolismo , Porcinos , Factores de Transcripción/genética , Proteínas no Estructurales Virales/metabolismo , Compartimentos de Replicación Viral/metabolismo
10.
Proc Natl Acad Sci U S A ; 116(37): 18571-18577, 2019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31375630

RESUMEN

Bacteriophage (phage) have attractive advantages as delivery systems compared with mammalian viruses, but have been considered poor vectors because they lack evolved strategies to confront and overcome mammalian cell barriers to infective agents. We reasoned that improved efficacy of delivery might be achieved through structural modification of the viral capsid to avoid pre- and postinternalization barriers to mammalian cell transduction. We generated multifunctional hybrid adeno-associated virus/phage (AAVP) particles to enable simultaneous display of targeting ligands on the phage's minor pIII proteins and also degradation-resistance motifs on the very numerous pVIII coat proteins. This genetic strategy of directed evolution bestows a next-generation of AAVP particles that feature resistance to fibrinogen adsorption or neutralizing antibodies and ability to escape endolysosomal degradation. This results in superior gene transfer efficacy in vitro and also in preclinical mouse models of rodent and human solid tumors. Thus, the unique functions of our next-generation AAVP particles enable improved targeted gene delivery to tumor cells.


Asunto(s)
Bacteriófago M13/genética , Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Neoplasias/terapia , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Bacteriófago M13/inmunología , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Línea Celular Tumoral , Dependovirus/inmunología , Endosomas/inmunología , Endosomas/virología , Vectores Genéticos/administración & dosificación , Vectores Genéticos/inmunología , Humanos , Lisosomas/inmunología , Lisosomas/virología , Ratones , Neoplasias/genética , Oligopéptidos/genética , Oligopéptidos/inmunología , Prueba de Estudio Conceptual , Ratas , Transducción Genética/métodos , Internalización del Virus , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Virol ; 94(14)2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32376619

RESUMEN

HIV-1 assembly occurs principally at the plasma membrane (PM) of infected cells. Gag polyprotein precursors (Pr55Gag) are targeted to the PM, and their binding is mediated by the interaction of myristoylated matrix domain and a PM-specific phosphoinositide, the phosphatidylinositol-(4,5)-bisphosphate [PI(4,5)P2]. The major synthesis pathway of PI(4,5)P2 involves the activity of phosphatidylinositol-4-phosphate 5-kinase family type 1 composed of three isoforms (PIP5K1α, PIP5K1ß, and PIP5K1γ). To examine whether the activity of a specific PIP5K1 isoform determines proper Pr55Gag localization at the PM, we compared the cellular behavior of Pr55Gag in the context of PIP5K1 inhibition using siRNAs that individually targeted each of the three isoforms in TZM-bl HeLa cells. We found that downregulation of PIP5K1α and PIP5K1γ strongly impaired the targeting of Pr55Gag to the PM with a rerouting of the polyprotein within intracellular compartments. The efficiency of Pr55Gag release was thus impaired through the silencing of these two isoforms, while PIP5K1ß is dispensable for Pr55Gag targeting to the PM. The PM mistargeting due to the silencing of PIP5K1α leads to Pr55Gag hydrolysis through lysosome and proteasome pathways, while the silencing of PIP5K1γ leads to Pr55Gag accumulation in late endosomes. Our findings demonstrated that, within the PIP5K1 family, only the PI(4,5)P2 pools produced by PIP5K1α and PIP5K1γ are involved in the Pr55Gag PM targeting process.IMPORTANCE PM specificity of Pr55Gag membrane binding is mediated through the interaction of PI(4,5)P2 with the matrix (MA) basic residues. It was shown that overexpression of a PI(4,5)P2-depleting enzyme strongly impaired PM localization of Pr55Gag However, cellular factors that control PI(4,5)P2 production required for Pr55Gag-PM targeting have not yet been characterized. In this study, by individually inhibiting PIP5K1 isoforms, we elucidated a correlation between PI(4,5)P2 metabolism pathways mediated by PIP5K1 isoforms and the targeting of Pr55Gag to the PM of TZM-bl HeLa cells. Confocal microscopy analyses of cells depleted from PIP5K1α and PIP5K1γ show a rerouting of Pr55Gag to various intracellular compartments. Notably, Pr55Gag is degraded by the proteasome and/or by the lysosomes in PIP5K1α-depleted cells, while Pr55Gag is targeted to endosomal vesicles in PIP5K1γ-depleted cells. Thus, our results highlight, for the first time, the roles of PIP5K1α and PIP5K1γ as determinants of Pr55Gag targeting to the PM.


Asunto(s)
Membrana Celular/metabolismo , Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , VIH-1/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/biosíntesis , Precursores de Proteínas/metabolismo , Membrana Celular/genética , Membrana Celular/virología , Endosomas/genética , Endosomas/metabolismo , Endosomas/virología , VIH-1/genética , Células HeLa , Humanos , Lisosomas/genética , Lisosomas/metabolismo , Lisosomas/virología , Fosfatidilinositol 4,5-Difosfato/genética , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Precursores de Proteínas/genética , Proteolisis
12.
J Virol ; 94(12)2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32295904

RESUMEN

Mumps virus (MuV), an enveloped RNA virus of the Paramyxoviridae family and the causative agent of mumps, affects the salivary glands and other glandular tissues as well as the central nervous system. The virus enters the cell by inducing the fusion of its envelope with the plasma membrane of the target cell. Membrane fusion is mediated by MuV envelope proteins: the hemagglutinin-neuraminidase and fusion (F) protein. Cleavage of the MuV F protein (MuV-F) into two subunits by the cellular protease furin is a prerequisite for fusion and virus infectivity. Here, we show that 293T (a derivative of HEK293) cells do not produce syncytia upon expression of MuV envelope proteins or MuV infection. This failure is caused by the inefficient MuV-F cleavage despite the presence of functional furin in 293T cells. An expression cloning strategy revealed that overexpression of lysosome-associated membrane proteins (LAMPs) confers on 293T cells the ability to produce syncytia upon expression of MuV envelope proteins. The LAMP family comprises the ubiquitously expressed LAMP1 and LAMP2, the interferon-stimulated gene product LAMP3, and the cell type-specific proteins. The expression level of the LAMP3 gene, but not of LAMP1 and LAMP2 genes, differed markedly between 293T and HEK293 cells. Overexpression of LAMP1, LAMP2, or LAMP3 allowed 293T cells to process MuV-F efficiently. Furthermore, these LAMPs were found to interact with both MuV-F and furin. Our results indicate that LAMPs support the furin-mediated cleavage of MuV-F and that, among them, LAMP3 may be critical for the process, at least in certain cells.IMPORTANCE The cellular protease furin mediates proteolytic cleavage of many host and pathogen proteins and plays an important role in viral envelope glycoprotein maturation. MuV, an enveloped RNA virus of the Paramyxoviridae family and an important human pathogen, enters the cell through the fusion of its envelope with the plasma membrane of the target cell. Membrane fusion is mediated by the viral attachment protein and the F protein. Cleavage of MuV-F into two subunits by furin is a prerequisite for fusion and virus infectivity. Here, we show that LAMPs support the furin-mediated cleavage of MuV-F. Expression levels of LAMPs affect the processing of MuV-F and MuV-mediated membrane fusion. Among LAMPs, the interferon-stimulated gene product LAMP3 is most critical in certain cells. Our study provides potential targets for anti-MuV therapeutics.


Asunto(s)
Furina/genética , Interacciones Huésped-Patógeno/genética , Proteínas de Membrana de los Lisosomas/genética , Lisosomas/virología , Virus de la Parotiditis/genética , Proteínas de Neoplasias/genética , Proteínas Virales de Fusión/genética , Células A549 , Animales , Membrana Celular/metabolismo , Membrana Celular/virología , Chlorocebus aethiops , Furina/metabolismo , Regulación de la Expresión Génica , Células Gigantes/química , Células Gigantes/metabolismo , Células HEK293 , Proteína HN/genética , Proteína HN/metabolismo , Células HeLa , Humanos , Proteína 2 de la Membrana Asociada a los Lisosomas/genética , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Proteínas de Membrana de los Lisosomas/metabolismo , Lisosomas/metabolismo , Virus de la Parotiditis/metabolismo , Proteínas de Neoplasias/metabolismo , Unión Proteica , Proteolisis , Transducción de Señal , Células Vero , Proteínas Virales de Fusión/metabolismo , Internalización del Virus
13.
PLoS Pathog ; 15(8): e1007955, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31461509

RESUMEN

Most intracellular pathogens that reside in a vacuole prevent transit of their compartment to lysosomal organelles. Effector mechanisms induced by the pro-inflammatory cytokine Interferon-gamma (IFNγ) can promote the delivery of pathogen-occupied vacuoles to lysosomes for proteolytic degradation and are therefore important for host defense against intracellular pathogens. The bacterial pathogen Coxiella burnetii is unique in that, transport to the lysosome is essential for replication. The bacterium modulates membrane traffic to create a specialized autophagolysosomal compartment called the Coxiella-containing vacuole (CCV). Importantly, IFNγ signaling inhibits intracellular replication of C. burnetii, raising the question of which IFNγ-activated mechanisms restrict replication of a lysosome-adapted pathogen. To address this question, siRNA was used to silence a panel of IFNγ-induced genes in HeLa cells to identify genes required for restriction of C. burnetii intracellular replication. This screen demonstrated that Indoleamine 2,3-dioxygenase 1 (IDO1) contributes to IFNγ-mediated restriction of C. burnetii. IDO1 is an enzyme that catabolizes cellular tryptophan to kynurenine metabolites thereby reducing tryptophan availability in cells. Cells deficient in IDO1 function were more permissive for C. burnetii replication when treated with IFNγ, and supplementing IFNγ-treated cells with tryptophan enhanced intracellular replication. Additionally, ectopic expression of IDO1 in host cells was sufficient to restrict replication of C. burnetii in the absence of IFNγ signaling. Using differentiated THP1 macrophage-like cells it was determined that IFNγ-activation resulted in IDO1 production, and that supplementation of IFNγ-activated THP1 cells with tryptophan enhanced C. burnetii replication. Thus, this study identifies IDO1 production as a key cell-autonomous defense mechanism that limits infection by C. burnetii, which suggests that peptides derived from hydrolysis of proteins in the CCV do not provide an adequate supply of tryptophan for bacterial replication.


Asunto(s)
Coxiella burnetii/patogenicidad , Interacciones Huésped-Patógeno , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Lisosomas/virología , Fiebre Q/prevención & control , ARN Interferente Pequeño/genética , Replicación Viral/genética , Coxiella burnetii/efectos de los fármacos , Células HeLa , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Interferón gamma/farmacología , Macrófagos/metabolismo , Macrófagos/patología , Macrófagos/virología , Fiebre Q/genética , Fiebre Q/virología , Triptófano/metabolismo
14.
FASEB J ; 34(6): 7253-7264, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32367579

RESUMEN

Drug repurposing is potentially the fastest available option in the race to identify safe and efficacious drugs that can be used to prevent and/or treat COVID-19. By describing the life cycle of the newly emergent coronavirus, SARS-CoV-2, in light of emerging data on the therapeutic efficacy of various repurposed antimicrobials undergoing testing against the virus, we highlight in this review a possible mechanistic convergence between some of these tested compounds. Specifically, we propose that the lysosomotropic effects of hydroxychloroquine and several other drugs undergoing testing may be responsible for their demonstrated in vitro antiviral activities against COVID-19. Moreover, we propose that Niemann-Pick disease type C (NPC), a lysosomal storage disorder, may provide new insights into potential future therapeutic targets for SARS-CoV-2, by highlighting key established features of the disorder that together result in an "unfavorable" host cellular environment that may interfere with viral propagation. Our reasoning evolves from previous biochemical and cell biology findings related to NPC, coupled with the rapidly evolving data on COVID-19. Our overall aim is to suggest that pharmacological interventions targeting lysosomal function in general, and those particularly capable of reversibly inducing transient NPC-like cellular and biochemical phenotypes, constitute plausible mechanisms that could be used to therapeutically target COVID-19.


Asunto(s)
Antivirales/farmacocinética , Betacoronavirus/fisiología , Infecciones por Coronavirus/tratamiento farmacológico , Reposicionamiento de Medicamentos , Endosomas/virología , Hidroxicloroquina/farmacología , Lisosomas/virología , Enfermedad de Niemann-Pick Tipo C/patología , Neumonía Viral/tratamiento farmacológico , Proteína ADAM17/fisiología , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/uso terapéutico , Alanina/análogos & derivados , Alanina/farmacología , Alanina/uso terapéutico , Enzima Convertidora de Angiotensina 2 , Antivirales/farmacología , Antivirales/uso terapéutico , Bencilisoquinolinas/farmacología , Bencilisoquinolinas/uso terapéutico , Transporte Biológico , COVID-19 , Catepsina L/fisiología , Endocitosis , Endosomas/efectos de los fármacos , Endosomas/fisiología , Glicopéptidos/farmacología , Glicopéptidos/uso terapéutico , Humanos , Hidroxicloroquina/farmacocinética , Hidroxicloroquina/uso terapéutico , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/fisiología , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Lípidos de la Membrana/metabolismo , Microdominios de Membrana/fisiología , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/metabolismo , Oxiesteroles/metabolismo , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Receptores Virales/metabolismo , SARS-CoV-2 , Serina Endopeptidasas/fisiología , Triazoles/farmacología , Triazoles/uso terapéutico , Internalización del Virus/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
15.
Proc Natl Acad Sci U S A ; 115(39): E9041-E9050, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30190430

RESUMEN

Gold nanoparticles (NPs) wrapped in a membrane can be utilized as artificial virus nanoparticles (AVNs) that combine the large nonblinking or bleaching optical cross-section of the NP core with the biological surface properties and functionalities provided by a self-assembled lipid membrane. We used these hybrid nanomaterials to test the roles of monosialodihexosylganglioside (GM3) and phosphatidylserine (PS) for a lipid-mediated targeting of virus-containing compartments (VCCs) in macrophages. GM3-presenting AVNs bind to CD169 (Siglec-1)-expressing macrophages, but inclusion of PS in the GM3-containing AVN membrane decreases binding. Molecular dynamics simulations of the AVN membrane and experimental binding studies of CD169 to GM3-presenting AVNs reveal Na+-mediated interactions between GM3 and PS as a potential cause of the antagonistic action on binding by the two negatively charged lipids. GM3-functionalized AVNs with no or low PS content localize to tetherin+, CD9+ VCC in a membrane composition-depending fashion, but increasing amounts of PS in the AVN membrane redirect the NP to lysosomal compartments. Interestingly, this compartmentalization is highly GM3 specific. Even AVNs presenting the related monosialotetrahexosylganglioside (GM1) fail to achieve an accumulation in VCC. AVN localization to VCC was observed for AVN with gold NP core but not for liposomes, suggesting that NP sequestration into VCC has additional requirements beyond ligand (GM3)-receptor (CD169) recognition that are related to the physical properties of the NP core. Our results confirm AVN as a scalable platform for elucidating the mechanisms of lipid-mediated viral entry pathways and for selective intracellular targeting.


Asunto(s)
Gangliósido G(M3)/metabolismo , Oro , Macrófagos/metabolismo , Membranas Artificiales , Nanopartículas del Metal , Fosfatidilserinas/metabolismo , Internalización del Virus , Virus/metabolismo , Humanos , Lisosomas/metabolismo , Lisosomas/virología , Macrófagos/virología , Lectina 1 Similar a Ig de Unión al Ácido Siálico/metabolismo , Células THP-1 , Tetraspanina 29/metabolismo
16.
Int J Mol Sci ; 22(4)2021 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-33670304

RESUMEN

Lysosomotropism is a biological characteristic of small molecules, independently present of their intrinsic pharmacological effects. Lysosomotropic compounds, in general, affect various targets, such as lipid second messengers originating from lysosomal enzymes promoting endothelial stress response in systemic inflammation; inflammatory messengers, such as IL-6; and cathepsin L-dependent viral entry into host cells. This heterogeneous group of drugs and active metabolites comprise various promising candidates with more favorable drug profiles than initially considered (hydroxy) chloroquine in prophylaxis and treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections/Coronavirus disease 2019 (COVID-19) and cytokine release syndrome (CRS) triggered by bacterial or viral infections. In this hypothesis, we discuss the possible relationships among lysosomotropism, enrichment in lysosomes of pulmonary tissue, SARS-CoV-2 infection, and transition to COVID-19. Moreover, we deduce further suitable approved drugs and active metabolites based with a more favorable drug profile on rational eligibility criteria, including readily available over-the-counter (OTC) drugs. Benefits to patients already receiving lysosomotropic drugs for other pre-existing conditions underline their vital clinical relevance in the current SARS-CoV2/COVID-19 pandemic.


Asunto(s)
Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Descubrimiento de Drogas , Lisosomas/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Antivirales/farmacocinética , Antivirales/uso terapéutico , COVID-19/inmunología , COVID-19/metabolismo , COVID-19/virología , Clorpromazina/farmacocinética , Clorpromazina/farmacología , Clorpromazina/uso terapéutico , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Descubrimiento de Drogas/métodos , Reposicionamiento de Medicamentos/métodos , Fluvoxamina/farmacocinética , Fluvoxamina/farmacología , Fluvoxamina/uso terapéutico , Humanos , Hidroxicloroquina/farmacocinética , Hidroxicloroquina/farmacología , Hidroxicloroquina/uso terapéutico , Interleucina-1/antagonistas & inhibidores , Interleucina-1/inmunología , Interleucina-6/antagonistas & inhibidores , Interleucina-6/inmunología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/virología , Lisosomas/inmunología , Lisosomas/metabolismo , Lisosomas/virología , SARS-CoV-2/inmunología , SARS-CoV-2/fisiología , Bibliotecas de Moléculas Pequeñas/farmacocinética , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Replicación Viral/efectos de los fármacos
17.
J Lipid Res ; 61(7): 972-982, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32457038

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus (SARS-CoV)-2 has resulted in the death of more than 328,000 persons worldwide in the first 5 months of 2020. Herculean efforts to rapidly design and produce vaccines and other antiviral interventions are ongoing. However, newly evolving viral mutations, the prospect of only temporary immunity, and a long path to regulatory approval pose significant challenges and call for a common, readily available, and inexpensive treatment. Strategic drug repurposing combined with rapid testing of established molecular targets could provide a pause in disease progression. SARS-CoV-2 shares extensive structural and functional conservation with SARS-CoV-1, including engagement of the same host cell receptor (angiotensin-converting enzyme 2) localized in cholesterol-rich microdomains. These lipid-enveloped viruses encounter the endosomal/lysosomal host compartment in a critical step of infection and maturation. Niemann-Pick type C (NP-C) disease is a rare monogenic neurodegenerative disease caused by deficient efflux of lipids from the late endosome/lysosome (LE/L). The NP-C disease-causing gene (NPC1) has been strongly associated with viral infection, both as a filovirus receptor (e.g., Ebola) and through LE/L lipid trafficking. This suggests that NPC1 inhibitors or NP-C disease mimetics could serve as anti-SARS-CoV-2 agents. Fortunately, there are such clinically approved molecules that elicit antiviral activity in preclinical studies, without causing NP-C disease. Inhibition of NPC1 may impair viral SARS-CoV-2 infectivity via several lipid-dependent mechanisms, which disturb the microenvironment optimum for viral infectivity. We suggest that known mechanistic information on NPC1 could be utilized to identify existing and future drugs to treat COVID-19.


Asunto(s)
Anticolesterolemiantes/uso terapéutico , Antivirales/uso terapéutico , Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Péptidos y Proteínas de Señalización Intracelular/genética , Enfermedad de Niemann-Pick Tipo C/tratamiento farmacológico , Pandemias , Neumonía Viral/tratamiento farmacológico , Androstenos/uso terapéutico , Enzima Convertidora de Angiotensina 2 , Betacoronavirus/metabolismo , Betacoronavirus/patogenicidad , COVID-19 , Colesterol/metabolismo , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/epidemiología , Reposicionamiento de Medicamentos/métodos , Humanos , Hidroxicloroquina/uso terapéutico , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Lisosomas/virología , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/genética , Enfermedad de Niemann-Pick Tipo C/metabolismo , Enfermedad de Niemann-Pick Tipo C/patología , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/diagnóstico , Neumonía Viral/epidemiología , Unión Proteica , Receptores Virales/antagonistas & inhibidores , Receptores Virales/genética , Receptores Virales/metabolismo , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo
18.
J Biol Chem ; 294(6): 2046-2059, 2019 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-30563842

RESUMEN

Insulin-induced gene 1 (INSIG1) regulates sterol synthesis by mediating the activation of sterol regulatory element-binding protein (SREBP) and the degradation of the HMG-CoA reductase (HMGCR). INSIG1 is up-regulated during HIV-1 infection, but its role in HIV-1 infection is unknown. In this report, using pseudovirus production, protein overexpression, and gene knockouts, we found that INSIG1 inhibits HIV-1 production by accelerating the degradation of the HIV-1 Gag protein. Unlike the degradation of HMGCR via the E3 ubiquitin ligase autocrine motility factor receptor (AMFR), a process that depends on the proteasome, INSIG1 coordinated with another ligase, translocation in renal carcinoma chromosome 8 (TRC8), and promoted Gag degradation through the lysosome pathway. We conclude that INSIG1 functions as a sentinel responsive to HIV-1 production and inhibits HIV-1 replication by degrading Gag, a process occurring at intracellular membrane sites such as the endoplasmic reticulum and endosomes where both INSIG1 and Gag may be located.


Asunto(s)
VIH-1/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteolisis , Receptores de Superficie Celular/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Replicación Viral , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo , Células HEK293 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Células Jurkat , Lisosomas/genética , Lisosomas/metabolismo , Lisosomas/virología , Proteínas de la Membrana/genética , Receptores de Superficie Celular/genética , Células THP-1 , Ubiquitina-Proteína Ligasas/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética
19.
J Virol ; 94(1)2019 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-31597781

RESUMEN

The divergent clinical outcomes of human T cell leukemia virus type 1 (HTLV-1) and HTLV-2 infections have been attributed to functional differences in their antisense proteins. In contrast to HTLV-1 bZIP factor (HBZ), the role of the antisense protein of HTLV-2 (APH-2) in HTLV-2 infection is poorly understood. In previous studies, we identified the endosomal sorting complex required for transport 0 (ESCRT-0) subunit HRS as a novel interaction partner of APH-2 but not HBZ. HRS is a master regulator of endosomal protein sorting for lysosomal degradation and is hijacked by many viruses to promote replication. However, no studies to date have shown a link between HTLVs and HRS. In this study, we sought to characterize the interaction between HRS and APH-2 and to investigate the impact of HRS on the life cycle of HTLV-2. We confirmed a direct specific interaction between APH-2 and HRS and showed that the CC2 domain of HRS and the N-terminal domain of APH-2 mediate their interaction. We demonstrated that HRS recruits APH-2 to early endosomes, possibly furnishing an entry route into the endosomal/lysosomal pathway. We demonstrated that inhibition of this pathway using either bafilomycin or HRS overexpression substantially extends the half-life of APH-2 and stabilizes Tax2B expression levels. We found that HRS enhances Tax2B-mediated long terminal repeat (LTR) activation, while depletion of HRS enhances HTLV-2 production and release, indicating that HRS may have a negative impact on HTLV-2 replication. Overall, our study provides important new insights into the role of the ESCRT-0 HRS protein, and by extension the ESCRT machinery and the endosomal/lysosomal pathway, in HTLV-2 infection.IMPORTANCE While APH-2 is the only viral protein consistently expressed in infected carriers, its role in HTLV-2 infection is poorly understood. In this study, we characterized the interaction between the ESCRT-0 component HRS and APH-2 and explored the role of HRS in HTLV-2 replication. HRS is a master regulator of protein sorting for lysosomal degradation, a feature that is manipulated by several viruses to promote replication. Unexpectedly, we found that HRS targets APH-2 and possibly Tax2B for lysosomal degradation and has an overall negative impact on HTLV-2 replication and release. The negative impact of interactions between HTLV-2 regulatory proteins and HRS, and by extension the ESCRT machinery, may represent an important strategy used by HTLV-2 to limit virus production and to promote persistence, features that may contribute to the limited pathogenic potential of this infection.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Productos del Gen tax/genética , Virus Linfotrópico T Tipo 2 Humano/genética , Fosfoproteínas/genética , Proteínas de los Retroviridae/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Cicloheximida/farmacología , Complejos de Clasificación Endosomal Requeridos para el Transporte/antagonistas & inhibidores , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Regulación de la Expresión Génica , Productos del Gen tax/metabolismo , Células HEK293 , Células HeLa , Virus Linfotrópico T Tipo 1 Humano/efectos de los fármacos , Virus Linfotrópico T Tipo 1 Humano/genética , Virus Linfotrópico T Tipo 1 Humano/metabolismo , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Virus Linfotrópico T Tipo 2 Humano/efectos de los fármacos , Virus Linfotrópico T Tipo 2 Humano/metabolismo , Virus Linfotrópico T Tipo 2 Humano/patogenicidad , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Lisosomas/virología , Macrólidos/farmacología , Fosfoproteínas/metabolismo , Proteínas de los Retroviridae/metabolismo , Transducción de Señal , Replicación Viral/efectos de los fármacos
20.
J Neurovirol ; 25(2): 263-274, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30746609

RESUMEN

Accelerated neurological disorders are increasingly prominent among the HIV-infected population and are likely driven by the toxicity from long-term use of antiretroviral drugs. We explored potential side effects of antiretroviral drugs in HIV-infected primary human astrocytes and whether opioid co-exposure exacerbates the response. HIV-infected human astrocytes were exposed to the reverse transcriptase inhibitor, emtricitabine, alone or in combination with two protease inhibitors ritonavir and atazanavir (ERA) with and without morphine co-exposure. The effect of the protease inhibitor, lopinavir, alone or in combination with the protease inhibitor, abacavir, and the integrase inhibitor, raltegravir (LAR), with and without morphine co-exposure was also explored. Exposure with emtricitabine alone or ERA in HIV-infected astrocytes caused a significant decrease in viral replication and attenuated HIV-induced inflammatory molecules, while co-exposure with morphine negated the inhibitory effects of ERA, leading to increased viral replication and inflammatory molecules. Exposure with emtricitabine alone or in combination with morphine caused a significant disruption of mitochondrial membrane integrity. Genetic analysis revealed a significant increase in the expression of p62/SQSTM1 which correlated with an increase in the histone-modifying enzyme, ESCO2, after exposure with ERA alone or in combination with morphine. Furthermore, several histone-modifying enzymes such as CIITA, PRMT8, and HDAC10 were also increased with LAR exposure alone or in combination with morphine. Accumulation of p62/SQSTM1 is indicative of dysfunctional lysosomal fusion. Together with the loss of mitochondrial integrity and epigenetic changes, these effects may lead to enhanced viral titer and inflammatory molecules contributing to the neuropathology associated with HIV.


Asunto(s)
Fármacos Anti-VIH/farmacología , Astrocitos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Morfina/efectos adversos , Narcóticos/efectos adversos , Proteína Sequestosoma-1/genética , Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Astrocitos/metabolismo , Astrocitos/virología , Sulfato de Atazanavir/farmacología , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Didesoxinucleósidos/farmacología , Combinación de Medicamentos , Emtricitabina/farmacología , VIH-1/efectos de los fármacos , VIH-1/crecimiento & desarrollo , VIH-1/patogenicidad , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Humanos , Lopinavir/farmacología , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Lisosomas/virología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/virología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Cultivo Primario de Células , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Raltegravir Potásico/farmacología , Ritonavir/farmacología , Proteína Sequestosoma-1/agonistas , Proteína Sequestosoma-1/metabolismo , Transducción de Señal , Transactivadores/genética , Transactivadores/metabolismo , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA