Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Virol ; 97(4): e0014423, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: covidwho-2297692

RESUMEN

2019 coronavirus disease (COVID-19) is a disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In addition to respiratory illness, COVID-19 patients exhibit neurological symptoms lasting from weeks to months (long COVID). It is unclear whether these neurological manifestations are due to an infection of brain cells. We found that a small fraction of human induced pluripotent stem cell (iPSC)-derived neurons, but not astrocytes, were naturally susceptible to SARS-CoV-2. Based on the inhibitory effect of blocking antibodies, the infection seemed to depend on the receptor angiotensin-converting enzyme 2 (ACE2), despite very low levels of its expression in neurons. The presence of double-stranded RNA in the cytoplasm (the hallmark of viral replication), abundant synthesis of viral late genes localized throughout infected cells, and an increase in the level of viral RNA in the culture medium (viral release) within the first 48 h of infection suggested that the infection was productive. Productive entry of SARS-CoV-2 requires the fusion of the viral and cellular membranes, which results in the delivery of the viral genome into the cytoplasm of the target cell. The fusion is triggered by proteolytic cleavage of the viral surface spike protein, which can occur at the plasma membrane or from endosomes or lysosomes. We found that SARS-CoV-2 infection of human neurons was insensitive to nafamostat and camostat, which inhibit cellular serine proteases, including transmembrane serine protease 2 (TMPRSS2). Inhibition of cathepsin L also did not significantly block infection. In contrast, the neuronal infection was blocked by apilimod, an inhibitor of phosphatidyl-inositol 5 kinase (PIK5K), which regulates early to late endosome maturation. IMPORTANCE COVID-19 is a disease caused by the coronavirus SARS-CoV-2. Millions of patients display neurological symptoms, including headache, impairment of memory, seizures, and encephalopathy, as well as anatomical abnormalities, such as changes in brain morphology. SARS-CoV-2 infection of the human brain has been documented, but it is unclear whether the observed neurological symptoms are linked to direct brain infection. The mechanism of virus entry into neurons has also not been characterized. Here, we investigated SARS-CoV-2 infection by using a human iPSC-derived neural cell model and found that a small fraction of cortical-like neurons was naturally susceptible to infection. The productive infection was ACE2 dependent and TMPRSS2 independent. We also found that the virus used the late endosomal and lysosomal pathway for cell entry and that the infection could be blocked by apilimod, an inhibitor of cellular PIK5K.


Asunto(s)
COVID-19 , Células Madre Pluripotentes Inducidas , SARS-CoV-2 , Humanos , Enzima Convertidora de Angiotensina 2 , COVID-19/fisiopatología , Endosomas/metabolismo , Endosomas/virología , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/metabolismo , Neuronas/virología , Síndrome Post Agudo de COVID-19/fisiopatología , Síndrome Post Agudo de COVID-19/virología , SARS-CoV-2/fisiología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus/efectos de los fármacos , Fosfotransferasas/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Astrocitos/virología , Células Cultivadas
3.
J Virol ; 96(17): e0077222, 2022 09 14.
Artículo en Inglés | MEDLINE | ID: covidwho-1992939

RESUMEN

Bats are reservoirs for diverse coronaviruses, including swine acute diarrhea syndrome coronavirus (SADS-CoV). SADS-CoV was first identified in diarrheal piglets in 2017. As a novel alphacoronavirus, SADS-CoV shares ~95% identity with bat alphacoronavirus HKU2. SADS-CoV has been reported to have broad cell tropism and inherent potential to cross host species barriers for dissemination. Thus far, no effective antiviral drugs or vaccines are available to treat infections with SADS-CoV. Therefore, knowledge of the protein-coding gene set and a subcellular localization map of SADS-CoV proteins are fundamental first steps in this endeavor. Here, all SADS-CoV genes were cloned separately into Flag-tagged plasmids, and the subcellular localizations of viral proteins, with the exception of nsp11, were detected using confocal microscopy techniques. As a result, nsp1, nsp3-N, nsp4, nsp5, nsp7, nsp8, nsp9, nsp10, nsp14, and nsp15 were localized in the cytoplasm and nuclear spaces, and these viral proteins may perform specific functions in the nucleus. All structural and accessory proteins were mainly localized in the cytoplasm. NS7a and membrane protein M colocalized with the Golgi compartment, and they may regulate the assembly of SADS-CoV virions. Maturation of SADS-CoV may occur in the late endosomes, during which envelope protein E is involved in the assembly and release of the virus. In summary, the present study demonstrates for the first time the location of all the viral proteins of SADS-CoV. These fundamental studies of SADS-CoV will promote studies of basic virology of SADS-CoV and support preventive strategies for animals with infection of SADS-CoV. IMPORTANCE SADS-CoV is the first documented spillover of a bat coronavirus that causes severe diseases in domestic animals. Our study is an in-depth annotation of the newly discovered swine coronavirus SADS-CoV genome and viral protein expression. Systematic subcellular localization of SADS-CoV proteins can have dramatic significance in revealing viral protein biological functions in the subcellular locations. Furthermore, our study promote understanding the fundamental science behind the novel swine coronavirus to pave the way for treatments and cures.


Asunto(s)
Alphacoronavirus , Infecciones por Coronavirus , Enfermedades de los Porcinos , Proteínas Virales , Alphacoronavirus/genética , Animales , Núcleo Celular/virología , Quirópteros , Infecciones por Coronavirus/veterinaria , Endosomas/virología , Aparato de Golgi/virología , Porcinos , Enfermedades de los Porcinos/virología , Proteínas Virales/genética
4.
Proc Natl Acad Sci U S A ; 119(25): e2201980119, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: covidwho-1890414

RESUMEN

Endosomal sorting maintains cellular homeostasis by recycling transmembrane proteins and associated proteins and lipids (termed "cargoes") from the endosomal network to multiple subcellular destinations, including retrograde traffic to the trans-Golgi network (TGN). Viral and bacterial pathogens subvert retrograde trafficking machinery to facilitate infectivity. Here, we develop a proteomic screen to identify retrograde cargo proteins of the endosomal SNX-BAR sorting complex promoting exit 1 (ESCPE-1). Using this methodology, we identify Neuropilin-1 (NRP1), a recently characterized host factor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, as a cargo directly bound and trafficked by ESCPE-1. ESCPE-1 mediates retrograde trafficking of engineered nanoparticles functionalized with the NRP1-interacting peptide of the SARS-CoV-2 spike (S) protein. CRISPR-Cas9 deletion of ESCPE-1 subunits reduces SARS-CoV-2 infection levels in cell culture. ESCPE-1 sorting of NRP1 may therefore play a role in the intracellular membrane trafficking of NRP1-interacting viruses such as SARS-CoV-2.


Asunto(s)
COVID-19 , Endosomas , Interacciones Huésped-Patógeno , Neuropilina-1 , SARS-CoV-2 , COVID-19/metabolismo , COVID-19/virología , Sistemas CRISPR-Cas , Endosomas/virología , Eliminación de Gen , Humanos , Nanopartículas , Neuropilina-1/genética , Neuropilina-1/metabolismo , Proteómica , SARS-CoV-2/metabolismo , Nexinas de Clasificación/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo
7.
Cell ; 184(24): 5950-5969.e22, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: covidwho-1499701

RESUMEN

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.


Asunto(s)
Autofagosomas/virología , COVID-19/virología , Autofagia , COVID-19/metabolismo , Sistemas CRISPR-Cas , Línea Celular Tumoral , Retículo Endoplásmico/metabolismo , Endosomas/fisiología , Endosomas/virología , Aparato de Golgi/fisiología , Células HEK293 , Células HeLa , Humanos , Fusión de Membrana , Microscopía Confocal , Fagosomas/metabolismo , Fagosomas/virología , Proteínas Qa-SNARE/biosíntesis , Receptores sigma/biosíntesis , SARS-CoV-2 , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/biosíntesis , Sinaptotagminas/biosíntesis
8.
J Cell Biochem ; 123(2): 155-160, 2022 02.
Artículo en Inglés | MEDLINE | ID: covidwho-1473858

RESUMEN

Drug repurposing is an attractive option for identifying new treatment strategies, in particular in extraordinary situations of urgent need such as the current coronavirus disease 2019 (Covid-19) pandemic. Recently, the World Health Organization announced testing of three drugs as potential Covid-19 therapeutics that are known for their dampening effect on the immune system. Thus, the underlying concept of selecting these drugs is to temper the potentially life-threatening overshooting of the immune system reacting to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection. This viewpoint discusses the possibility that the impact of these and other drugs on autophagy contributes to their therapeutic effect by hampering the SARS-CoV-2 life cycle.


Asunto(s)
Antivirales/farmacología , Artesunato/farmacología , Autofagia/efectos de los fármacos , Tratamiento Farmacológico de COVID-19 , Reposicionamiento de Medicamentos , Mesilato de Imatinib/farmacología , Infliximab/farmacología , Pandemias , SARS-CoV-2/efectos de los fármacos , Antidepresivos/farmacología , Antivirales/uso terapéutico , Artesunato/uso terapéutico , Cloroquina/farmacología , Desarrollo de Medicamentos , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/fisiología , Retículo Endoplásmico/virología , Endosomas/efectos de los fármacos , Endosomas/virología , Humanos , Hidroxicloroquina/farmacología , Mesilato de Imatinib/uso terapéutico , Infliximab/uso terapéutico , Membranas Intracelulares/efectos de los fármacos , Membranas Intracelulares/fisiología , Membranas Intracelulares/virología , Ivermectina/farmacología , Macrólidos/farmacología , Coronavirus del Síndrome Respiratorio de Oriente Medio/efectos de los fármacos , Niclosamida/farmacología , Niclosamida/uso terapéutico , ARN Viral/metabolismo , SARS-CoV-2/fisiología , Replicación Viral
9.
FEBS J ; 288(17): 5071-5088, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: covidwho-1393880

RESUMEN

While there is undeniable evidence to link endosomal acid-base homeostasis to viral pathogenesis, the lack of druggable molecular targets has hindered translation from bench to bedside. The recent identification of variants in the interferon-inducible endosomal Na+ /H+ exchanger 9 associated with severe coronavirus disease-19 (COVID-19) has brought a shift in the way we envision aberrant endosomal acidification. Is it linked to an increased susceptibility to viral infection or a propensity to develop critical illness? This review summarizes the genetic and cellular evidence linking endosomal Na+ /H+ exchangers and viral diseases to suggest how they can act as a broad-spectrum modulator of viral infection and downstream pathophysiology. The review also presents novel insights supporting the complex role of endosomal acid-base homeostasis in viral pathogenesis and discusses the potential causes for negative outcomes of clinical trials utilizing alkalinizing drugs as therapies for COVID-19. These findings lead to a pathogenic model of viral disease that predicts that nonspecific targeting of endosomal pH might fail, even if administered early on, and suggests that endosomal Na+ /H+ exchangers may regulate key host antiviral defence mechanisms and mediators that act to drive inflammatory organ injury.


Asunto(s)
COVID-19/terapia , SARS-CoV-2/patogenicidad , Intercambiadores de Sodio-Hidrógeno/genética , Virosis/terapia , COVID-19/genética , COVID-19/virología , Endosomas/genética , Endosomas/virología , Humanos , Protones , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Virosis/genética , Virosis/virología
10.
Antiviral Res ; 194: 105167, 2021 10.
Artículo en Inglés | MEDLINE | ID: covidwho-1370440

RESUMEN

Niemann-Pick type C1 (NPC1) receptor is an endosomal membrane protein that regulates intracellular cholesterol traffic. This protein has been shown to play an important role for several viruses. It has been reported that SARS-CoV-2 enters the cell through plasma membrane fusion and/or endosomal entry upon availability of proteases. However, the whole process is not fully understood yet and additional viral/host factors might be required for viral fusion and subsequent viral replication. Here, we report a novel interaction between the SARS-CoV-2 nucleoprotein (N) and the cholesterol transporter NPC1. Furthermore, we have found that some compounds reported to interact with NPC1, carbazole SC816 and sulfides SC198 and SC073, were able to reduce SARS-CoV-2 viral infection with a good selectivity index in human cell infection models. These findings suggest the importance of NPC1 for SARS-CoV-2 viral infection and a new possible potential therapeutic target to fight against COVID-19.


Asunto(s)
Transporte Biológico , Tratamiento Farmacológico de COVID-19 , Endosomas/virología , Proteína Niemann-Pick C1/análisis , SARS-CoV-2/fisiología , Animales , Carbazoles/farmacología , Chlorocebus aethiops , Endosomas/química , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Fusión de Membrana , Células Vero , Replicación Viral
11.
Methods Mol Biol ; 2099: 9-20, 2020.
Artículo en Inglés | MEDLINE | ID: covidwho-1292544

RESUMEN

Middle East respiratory syndrome coronavirus (MERS-CoV) is an emerging zoonotic pathogen with a broad host range. The extent of MERS-CoV in nature can be traced to its adaptable cell entry steps. The virus can bind host-cell carbohydrates as well as proteinaceous receptors. Following receptor interaction, the virus can utilize diverse host proteases for cleavage activation of virus-host cell membrane fusion and subsequent genome delivery. The fusion and genome delivery steps can be completed at variable times and places, either at or near cell surfaces or deep within endosomes. Investigators focusing on the CoVs have developed several methodologies that effectively distinguish these different cell entry pathways. Here we describe these methods, highlighting virus-cell entry factors, entry inhibitors, and viral determinants that specify the cell entry routes. While the specific methods described herein were utilized to reveal MERS-CoV entry pathways, they are equally suited for other CoVs, as well as other protease-dependent viral species.


Asunto(s)
Infecciones por Coronavirus/virología , Genoma Viral/genética , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Internalización del Virus , Membrana Celular/virología , Endosomas/virología , Células HEK293 , Humanos , Proteínas de la Membrana/metabolismo , Coronavirus del Síndrome Respiratorio de Oriente Medio/genética , Coronavirus del Síndrome Respiratorio de Oriente Medio/aislamiento & purificación , Péptido Hidrolasas/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptores Virales/genética , Receptores Virales/metabolismo , Serina Endopeptidasas/metabolismo , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo
12.
Cells ; 10(5)2021 05 07.
Artículo en Inglés | MEDLINE | ID: covidwho-1223961

RESUMEN

The flavonoid naringenin (Nar), present in citrus fruits and tomatoes, has been identified as a blocker of an emerging class of human intracellular channels, namely the two-pore channel (TPC) family, whose role has been established in several diseases. Indeed, Nar was shown to be effective against neoangiogenesis, a process essential for solid tumor progression, by specifically impairing TPC activity. The goal of the present review is to illustrate the rationale that links TPC channels to the mechanism of coronavirus infection, and how their inhibition by Nar could be an efficient pharmacological strategy to fight the current pandemic plague COVID-19.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/metabolismo , Flavanonas/farmacología , Neoplasias/tratamiento farmacológico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antivirales/farmacología , Antivirales/uso terapéutico , Arabidopsis/metabolismo , COVID-19/epidemiología , COVID-19/patología , COVID-19/virología , Bloqueadores de los Canales de Calcio/uso terapéutico , Evaluación Preclínica de Medicamentos , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Endosomas/virología , Flavanonas/uso terapéutico , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Lisosomas/virología , Neoplasias/irrigación sanguínea , Neoplasias/patología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Pandemias/prevención & control , SARS-CoV-2/patogenicidad , Vacuolas/metabolismo , Internalización del Virus/efectos de los fármacos
13.
J Biol Chem ; 296: 100306, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1152462

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of COVID-19, so understanding its biology and infection mechanisms is critical to facing this major medical challenge. SARS-CoV-2 is known to use its spike glycoprotein to interact with the cell surface as a first step in the infection process. As for other coronaviruses, it is likely that SARS-CoV-2 next undergoes endocytosis, but whether or not this is required for infectivity and the precise endocytic mechanism used are unknown. Using purified spike glycoprotein and lentivirus pseudotyped with spike glycoprotein, a common model of SARS-CoV-2 infectivity, we now demonstrate that after engagement with the plasma membrane, SARS-CoV-2 undergoes rapid, clathrin-mediated endocytosis. This suggests that transfer of viral RNA to the cell cytosol occurs from the lumen of the endosomal system. Importantly, we further demonstrate that knockdown of clathrin heavy chain, which blocks clathrin-mediated endocytosis, reduces viral infectivity. These discoveries reveal that SARS-CoV-2 uses clathrin-mediated endocytosis to gain access into cells and suggests that this process is a key aspect of virus infectivity.


Asunto(s)
Enzima Convertidora de Angiotensina 2/genética , Cadenas Pesadas de Clatrina/genética , Endocitosis/genética , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética , Internalización del Virus/efectos de los fármacos , Células A549 , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Chlorocebus aethiops , Cadenas Pesadas de Clatrina/antagonistas & inhibidores , Cadenas Pesadas de Clatrina/metabolismo , Endocitosis/efectos de los fármacos , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Endosomas/virología , Regulación de la Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Células HEK293 , Interacciones Huésped-Patógeno/genética , Humanos , Hidrazonas/farmacología , Lentivirus/genética , Lentivirus/metabolismo , Unión Proteica/efectos de los fármacos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/metabolismo , Transducción de Señal , Glicoproteína de la Espiga del Coronavirus/metabolismo , Sulfonamidas/farmacología , Tiazolidinas/farmacología , Células Vero
14.
Nat Commun ; 12(1): 961, 2021 02 11.
Artículo en Inglés | MEDLINE | ID: covidwho-1078585

RESUMEN

The global spread of SARS-CoV-2 is posing major public health challenges. One feature of SARS-CoV-2 spike protein is the insertion of multi-basic residues at the S1/S2 subunit cleavage site. Here, we find that the virus with intact spike (Sfull) preferentially enters cells via fusion at the plasma membrane, whereas a clone (Sdel) with deletion disrupting the multi-basic S1/S2 site utilizes an endosomal entry pathway. Using Sdel as model, we perform a genome-wide CRISPR screen and identify several endosomal entry-specific regulators. Experimental validation of hits from the CRISPR screen shows that host factors regulating the surface expression of angiotensin-converting enzyme 2 (ACE2) affect entry of Sfull virus. Animal-to-animal transmission with the Sdel virus is reduced compared to Sfull in the hamster model. These findings highlight the critical role of the S1/S2 boundary of SARS-CoV-2 spike protein in modulating virus entry and transmission and provide insights into entry of coronaviruses.


Asunto(s)
COVID-19/virología , Sistemas CRISPR-Cas , Estudio de Asociación del Genoma Completo , Interacciones Huésped-Patógeno , SARS-CoV-2/fisiología , Internalización del Virus , Células A549 , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , COVID-19/genética , Chlorocebus aethiops , Modelos Animales de Enfermedad , Endosomas/virología , Células HeLa , Humanos , Mesocricetus , Serina Endopeptidasas , Glicoproteína de la Espiga del Coronavirus/metabolismo , Células Vero
15.
Cell Calcium ; 94: 102360, 2021 03.
Artículo en Inglés | MEDLINE | ID: covidwho-1064903

RESUMEN

Ion channels are necessary for correct lysosomal function including degradation of cargoes originating from endocytosis. Almost all enveloped viruses, including coronaviruses (CoVs), enter host cells via endocytosis, and do not escape endosomal compartments into the cytoplasm (via fusion with the endolysosomal membrane) unless the virus-encoded envelope proteins are cleaved by lysosomal proteases. With the ongoing outbreak of severe acute respiratory syndrome (SARS)-CoV-2, endolysosomal two-pore channels represent an exciting and emerging target for antiviral therapies. This review focuses on the latest knowledge of the effects of lysosomal ion channels on the cellular entry and uncoating of enveloped viruses, which may aid in development of novel therapies against emerging infectious diseases such as SARS-CoV-2.


Asunto(s)
Antivirales/uso terapéutico , COVID-19/virología , Canales Iónicos/fisiología , Lisosomas/virología , SARS-CoV-2/fisiología , Envoltura Viral/fisiología , Internalización del Virus , Desencapsidación Viral , Aminopiridinas/farmacología , Aminopiridinas/uso terapéutico , Antivirales/farmacología , Diseño de Fármacos , Endocitosis , Endosomas/metabolismo , Endosomas/virología , Compuestos Heterocíclicos con 3 Anillos/farmacología , Compuestos Heterocíclicos con 3 Anillos/uso terapéutico , Humanos , Hidrazonas/farmacología , Hidrazonas/uso terapéutico , Canales Iónicos/clasificación , Lisosomas/enzimología , Lisosomas/metabolismo , Modelos Biológicos , Morfolinas/farmacología , Morfolinas/uso terapéutico , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , ATPasas de Translocación de Protón Vacuolares/fisiología , Internalización del Virus/efectos de los fármacos , Desencapsidación Viral/efectos de los fármacos
17.
Emerg Microbes Infect ; 9(1): 2245-2255, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: covidwho-795734

RESUMEN

The Coronavirus Disease 2019 (COVID-19) pandemic caused by the Severe Acute Respiratory Syndrome Related Coronavirus 2 (SARS-CoV-2) is a global health emergency. As only very limited therapeutic options are clinically available, there is an urgent need for the rapid development of safe, effective, and globally available pharmaceuticals that inhibit SARS-CoV-2 entry and ameliorate COVID-19 severity. In this study, we explored the use of small compounds acting on the homeostasis of the endolysosomal host-pathogen interface, to fight SARS-CoV-2 infection. We find that fluoxetine, a widely used antidepressant and a functional inhibitor of acid sphingomyelinase (FIASMA), efficiently inhibited the entry and propagation of SARS-CoV-2 in the cell culture model without cytotoxic effects and also exerted potent antiviral activity against two currently circulating influenza A virus subtypes, an effect which was also observed upon treatment with the FIASMAs amiodarone and imipramine. Mechanistically, fluoxetine induced both impaired endolysosomal acidification and the accumulation of cholesterol within the endosomes. As the FIASMA group consists of a large number of small compounds that are well-tolerated and widely used for a broad range of clinical applications, exploring these licensed pharmaceuticals may offer a variety of promising antivirals for host-directed therapy to counteract enveloped viruses, including SARS-CoV-2.


Asunto(s)
Antidepresivos/farmacología , Antivirales/farmacología , Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus/virología , Inhibidores Enzimáticos/farmacología , Fluoxetina/farmacología , Neumonía Viral/virología , Betacoronavirus/fisiología , COVID-19 , Línea Celular , Endosomas/virología , Humanos , Pandemias , SARS-CoV-2 , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos
18.
Viruses ; 12(7)2020 07 03.
Artículo en Inglés | MEDLINE | ID: covidwho-636175

RESUMEN

Chikungunya virus (CHIKV) is an enveloped virus that enters host cells and transits within the endosomes before starting its replication cycle, the precise mechanism of which is yet to be elucidated. Endocytosis and endosome acidification inhibitors inhibit infection by CHIKV, murine leukemia virus (MLV), or SARS-coronavirus, indicating that these viral entries into host cells occur through endosomes and require endosome acidification. Although endosomal cathepsin B protease is necessary for MLV, Ebola virus, and SARS-CoV infections, its role in CHIKV infection is unknown. Our results revealed that endocytosis inhibitors attenuated CHIKV-pseudotyped MLV vector infection in 293T cells but not in TE671 cells. In contrast, macropinocytosis inhibitors attenuated CHIKV-pseudotyped MLV vector infection in TE671 cells but not in 293T cells, suggesting that CHIKV host cell entry occurs via endocytosis or macropinocytosis, depending on the cell lines used. Cathepsin B inhibitor and knockdown by an shRNA suppressed CHIKV-pseudotyped MLV vector infection both in 293T and TE671 cells. These results show that cathepsin B facilitates CHIKV infection regardless of the entry pathway.


Asunto(s)
Catepsina B/metabolismo , Fiebre Chikungunya/patología , Virus Chikungunya/fisiología , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Catepsina B/antagonistas & inhibidores , Línea Celular Tumoral , Endocitosis/fisiología , Endosomas/virología , Células HEK293 , Células HeLa , Humanos , Virus de la Leucemia Murina/fisiología , Pinocitosis/fisiología , Interferencia de ARN , ARN Interferente Pequeño/genética
19.
Science ; 370(6513): 241-247, 2020 10 09.
Artículo en Inglés | MEDLINE | ID: covidwho-733186

RESUMEN

Recent outbreaks of Ebola virus (EBOV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have exposed our limited therapeutic options for such diseases and our poor understanding of the cellular mechanisms that block viral infections. Using a transposon-mediated gene-activation screen in human cells, we identify that the major histocompatibility complex (MHC) class II transactivator (CIITA) has antiviral activity against EBOV. CIITA induces resistance by activating expression of the p41 isoform of invariant chain CD74, which inhibits viral entry by blocking cathepsin-mediated processing of the Ebola glycoprotein. We further show that CD74 p41 can block the endosomal entry pathway of coronaviruses, including SARS-CoV-2. These data therefore implicate CIITA and CD74 in host defense against a range of viruses, and they identify an additional function of these proteins beyond their canonical roles in antigen presentation.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/fisiología , Betacoronavirus/fisiología , Infecciones por Coronavirus/inmunología , Ebolavirus/fisiología , Fiebre Hemorrágica Ebola/inmunología , Antígenos de Histocompatibilidad Clase II/fisiología , Interacciones Huésped-Patógeno/inmunología , Proteínas Nucleares/fisiología , Neumonía Viral/inmunología , Transactivadores/fisiología , Internalización del Virus , Antígenos de Diferenciación de Linfocitos B/genética , COVID-19 , Línea Celular Tumoral , Infecciones por Coronavirus/virología , Elementos Transponibles de ADN , Endosomas/virología , Pruebas Genéticas , Fiebre Hemorrágica Ebola/virología , Antígenos de Histocompatibilidad Clase II/genética , Interacciones Huésped-Patógeno/genética , Humanos , Proteínas Nucleares/genética , Pandemias , Neumonía Viral/virología , SARS-CoV-2 , Transactivadores/genética , Transcripción Genética
20.
Open Heart ; 7(1)2020 06.
Artículo en Inglés | MEDLINE | ID: covidwho-595177

RESUMEN

The high rate of thrombotic complications associated with COVID-19 seems likely to reflect viral infection of vascular endothelial cells, which express the ACE2 protein that enables SARS-CoV-2 to invade cells. Various proinflammatory stimuli can promote thrombosis by inducing luminal endothelial expression of tissue factor (TF), which interacts with circulating coagulation factor VII to trigger extrinsic coagulation. The signalling mechanism whereby these stimuli evoke TF expression entails activation of NADPH oxidase, upstream from activation of the NF-kappaB transcription factor that drives the induced transcription of the TF gene. When single-stranded RNA viruses are taken up into cellular endosomes, they stimulate endosomal formation and activation of NADPH oxidase complexes via RNA-responsive toll-like receptor 7. It is therefore proposed that SARS-CoV-2 infection of endothelial cells evokes the expression of TF which is contingent on endosomal NADPH oxidase activation. If this hypothesis is correct, hydroxychloroquine, spirulina (more specifically, its chromophore phycocyanobilin) and high-dose glycine may have practical potential for mitigating the elevated thrombotic risk associated with COVID-19.


Asunto(s)
Betacoronavirus/patogenicidad , Coagulación Sanguínea , Infecciones por Coronavirus/virología , Endosomas/virología , Células Endoteliales/virología , NADPH Oxidasas/metabolismo , Neumonía Viral/virología , Tromboplastina/metabolismo , Trombosis/virología , Animales , Antivirales/uso terapéutico , Betacoronavirus/efectos de los fármacos , Coagulación Sanguínea/efectos de los fármacos , COVID-19 , Infecciones por Coronavirus/sangre , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/enzimología , Endosomas/efectos de los fármacos , Endosomas/enzimología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Activación Enzimática , Fibrinolíticos/uso terapéutico , Interacciones Huésped-Patógeno , Humanos , Pandemias , Neumonía Viral/sangre , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/enzimología , SARS-CoV-2 , Transducción de Señal , Trombosis/sangre , Trombosis/enzimología , Trombosis/prevención & control , Tratamiento Farmacológico de COVID-19
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA