Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Chem Inf Model ; 64(13): 5262-5272, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38869471

RESUMEN

Venezuelan equine encephalitis virus (VEEV) is a highly virulent pathogen whose nuclear localization signal (NLS) sequence from capsid protein binds to the host importin-α transport protein and blocks nuclear import. We studied the molecular mechanisms by which two small ligands, termed I1 and I2, interfere with the binding of VEEV's NLS peptide to importin-α protein. To this end, we performed all-atom replica exchange molecular dynamics simulations probing the competitive binding of the VEEV coreNLS peptide and I1 or I2 ligand to the importin-α major NLS binding site. As a reference, we used our previous simulations, which examined noncompetitive binding of the coreNLS peptide or the inhibitors to importin-α. We found that both inhibitors completely abrogate the native binding of the coreNLS peptide, forcing it to adopt a manifold of nonnative loosely bound poses within the importin-α major NLS binding site. Both inhibitors primarily destabilize the native coreNLS binding by masking its amino acids rather than competing with it for binding to importin-α. Because I2, in contrast to I1, binds off-site localizing on the edge of the major NLS binding site, it inhibits fewer coreNLS native binding interactions than I1. Structural analysis is supported by computations of the free energies of the coreNLS peptide binding to importin-α with or without competition from the inhibitors. Specifically, both inhibitors reduce the free energy gain from coreNLS binding, with I1 causing significantly larger loss than I2. To test our simulations, we performed AlphaScreen experiments measuring IC50 values for both inhibitors. Consistent with in silico results, the IC50 value for I1 was found to be lower than that for I2. We hypothesize that the inhibitory action of I1 and I2 ligands might be specific to the NLS from VEEV's capsid protein.


Asunto(s)
Unión Competitiva , Simulación de Dinámica Molecular , Señales de Localización Nuclear , alfa Carioferinas , alfa Carioferinas/metabolismo , alfa Carioferinas/química , alfa Carioferinas/antagonistas & inhibidores , Ligandos , Señales de Localización Nuclear/química , Virus de la Encefalitis Equina Venezolana/metabolismo , Virus de la Encefalitis Equina Venezolana/química , Unión Proteica , Péptidos/química , Péptidos/metabolismo , Péptidos/farmacología , Secuencia de Aminoácidos
2.
Cells ; 11(7)2022 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-35406765

RESUMEN

The global burden of malaria and toxoplasmosis has been limited by the use of efficacious anti-parasitic agents, however, emerging resistance in Plasmodium species and Toxoplasma gondii threatens disease control worldwide, implying that new agents/therapeutic targets are urgently needed. Nuclear localization signal (NLS)-dependent transport into the nucleus, mediated by members of the importin (IMP) superfamily of nuclear transporters, has shown potential as a target for intervention to limit viral infection. Here, we show for the first time that IMPα from P. falciparum and T. gondii have promise as targets for small molecule inhibitors. We use high-throughput screening to identify agents able to inhibit P. falciparum IMPα binding to a P. falciparum NLS, identifying a number of compounds that inhibit binding in the µM-nM range, through direct binding to P. falciparum IMPα, as shown in thermostability assays. Of these, BAY 11-7085 is shown to be a specific inhibitor of P. falciparum IMPα-NLS recognition. Importantly, a number of the inhibitors limited growth by both P. falciparum and T. gondii. The results strengthen the hypothesis that apicomplexan IMPα proteins have potential as therapeutic targets to aid in identifying novel agents for two important, yet neglected, parasitic diseases.


Asunto(s)
Plasmodium falciparum , alfa Carioferinas , Ensayos Analíticos de Alto Rendimiento , Señales de Localización Nuclear/metabolismo , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/metabolismo , Unión Proteica , alfa Carioferinas/antagonistas & inhibidores
3.
ChemMedChem ; 17(1): e202100640, 2022 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-34623765

RESUMEN

Taspase1 is a unique protease not only pivotal for embryonic development but also implicated in leukemia as well as solid tumors. As such, it is a promising target in cancer therapy, although only a limited number of Taspase1 inhibitors lacking general applicability are currently available. Here we present a bivalent guanidiniocarbonyl-pyrrole (GCP)-containing supramolecular ligand that is capable of disrupting the essential interaction between Taspase1 and its cognate import receptor Importin α in a concentration-dependent manner in vitro with an IC50 of 35 µM. Here, size of the bivalent vs the monovalent construct as well as its derivation with an aromatic cbz-group arose as critical determinants for efficient interference of 2GC. This was also evident when we investigated the effects in different tumor cell lines, resulting in comparable EC50 values (∼40-70 µM). Of note, in higher concentrations, 2GC also interfered with Taspase1's proteolytic activity. We thus believe to set the stage for a novel class of Taspase1 inhibitors targeting a pivotal protein-protein interaction prerequisite for its cancer-associated proteolytic function.


Asunto(s)
Endopeptidasas/metabolismo , Guanidina/farmacología , Inhibidores de Proteasas/farmacología , Pirroles/farmacología , alfa Carioferinas/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Endopeptidasas/química , Guanidina/química , Humanos , Ligandos , Estructura Molecular , Inhibidores de Proteasas/química , Pirroles/química , Relación Estructura-Actividad , alfa Carioferinas/química , alfa Carioferinas/metabolismo
4.
Biophys Chem ; 278: 106677, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34428682

RESUMEN

The SARS-CoV-2 pandemic has accelerated the study of existing drugs. The mixture of homologs called ivermectin (avermectin-B1a [HB1a] + avermectin-B1b [HB1b]) has shown antiviral activity against SARS-CoV-2 in vitro. However, there are few reports on the behavior of each homolog. We investigated the interaction of each homolog with promising targets of interest associated with SARS-CoV-2 infection from a biophysical and computational-chemistry perspective using docking and molecular dynamics. We observed a differential behavior for each homolog, with an affinity of HB1b for viral structures, and of HB1a for host structures considered. The induced disturbances were differential and influenced by the hydrophobicity of each homolog and of the binding pockets. We present the first comparative analysis of the potential theoretical inhibitory effect of both avermectins on biomolecules associated with COVID-19, and suggest that ivermectin through its homologs, has a multiobjective behavior.


Asunto(s)
Antivirales/química , Proteasas 3C de Coronavirus/antagonistas & inhibidores , ADN Helicasas/antagonistas & inhibidores , Ivermectina/análogos & derivados , alfa Carioferinas/antagonistas & inhibidores , beta Carioferinas/antagonistas & inhibidores , Animales , Antivirales/farmacología , Sitios de Unión , COVID-19/virología , Proteasas 3C de Coronavirus/química , Proteasas 3C de Coronavirus/metabolismo , ADN Helicasas/química , ADN Helicasas/metabolismo , Humanos , Ivermectina/química , Ivermectina/farmacología , Cinética , Ratones , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , SARS-CoV-2/química , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/enzimología , Termodinámica , alfa Carioferinas/química , alfa Carioferinas/metabolismo , beta Carioferinas/química , beta Carioferinas/metabolismo , Tratamiento Farmacológico de COVID-19
5.
Front Immunol ; 12: 663586, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33859652

RESUMEN

As of January 2021, SARS-CoV-2 has killed over 2 million individuals across the world. As such, there is an urgent need for vaccines and therapeutics to reduce the burden of COVID-19. Several vaccines, including mRNA, vector-based vaccines, and inactivated vaccines, have been approved for emergency use in various countries. However, the slow roll-out of vaccines and insufficient global supply remains a challenge to turn the tide of the pandemic. Moreover, vaccines are important tools for preventing the disease but therapeutic tools to treat patients are also needed. As such, since the beginning of the pandemic, repurposed FDA-approved drugs have been sought as potential therapeutic options for COVID-19 due to their known safety profiles and potential anti-viral effects. One of these drugs is ivermectin (IVM), an antiparasitic drug created in the 1970s. IVM later exerted antiviral activity against various viruses including SARS-CoV-2. In this review, we delineate the story of how this antiparasitic drug was eventually identified as a potential treatment option for COVID-19. We review SARS-CoV-2 lifecycle, the role of the nucleocapsid protein, the turning points in past research that provided initial 'hints' for IVM's antiviral activity and its molecular mechanism of action- and finally, we culminate with the current clinical findings.


Asunto(s)
Transporte Activo de Núcleo Celular/efectos de los fármacos , Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Ivermectina/uso terapéutico , SARS-CoV-2/efectos de los fármacos , Animales , Línea Celular , Chlorocebus aethiops , Proteínas de la Nucleocápside de Coronavirus/antagonistas & inhibidores , Proteínas de la Nucleocápside de Coronavirus/metabolismo , Reposicionamiento de Medicamentos , Humanos , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/metabolismo , Transporte de Proteínas/efectos de los fármacos , SARS-CoV-2/crecimiento & desarrollo , Células Vero , Replicación Viral/efectos de los fármacos , alfa Carioferinas/antagonistas & inhibidores , beta Carioferinas/antagonistas & inhibidores
6.
Biochem Pharmacol ; 186: 114501, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33684389

RESUMEN

Lactucopicrin, a bitter sesquiterpene lactone of leafy vegetables, such as chicory, curly escarole, and lettuce, possesses anti-malarial, anti-cancer and analgesic properties. However, it remains unknown whether lactucopicrin could inhibit vascular endothelial nuclear factor-κB (NF-κB) activation, a hallmark of vascular inflammatory diseases including sepsis. In tumor necrosis factor-α-stimulated human or mouse aortic endothelial cells, lactucopicrin dose-dependently inhibited NF-κB activation, and concomitantly repressed both vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1)-mediated monocyte adhesion. The lactucopicrin effect was not due to modulation of inhibitor of NF-κB kinases (IKK) α/ß/γ, inhibitor of NF-κB alpha (IκBα), and NF-κB/p65 DNA binding activity. Instead, lactucopicrin inhibited importin-α3 expression by destabilization of its mRNA, an effect mediating the lactucopicrin effect on NF-κB activity. More importantly, in lipopolysaccharide (LPS)-elicited septic mice, oral gavage with lactucopicrin decreased mortality by 30.5% as compared with the control treatment. This effect was associated with inhibited importin-α3 expression, suppressed NF-κB activation and VCAM-1/ICAM-1 expression, and inhibited leukocyte influx in the vascular endothelium of both lung and aorta. Collectively, our novel data suggest that dietary supplementation with lactucopicrin inhibits endothelial NF-κB activation by down-regulation of importin-α3 and thereby improves sepsis.


Asunto(s)
Células Endoteliales/metabolismo , Lactonas/uso terapéutico , FN-kappa B/metabolismo , Sepsis/tratamiento farmacológico , Sepsis/metabolismo , Sesquiterpenos/uso terapéutico , alfa Carioferinas/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células HL-60 , Humanos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Lactonas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Sepsis/inducido químicamente , Sesquiterpenos/farmacología , alfa Carioferinas/antagonistas & inhibidores
7.
Biochem Biophys Res Commun ; 538: 163-172, 2021 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-33341233

RESUMEN

FDA approved for parasitic indications, the small molecule ivermectin has been the focus of growing attention in the last 8 years due to its potential as an antiviral. We first identified ivermectin in a high throughput compound library screen as an agent potently able to inhibit recognition of the nuclear localizing Human Immunodeficiency Virus-1 (HIV-1) integrase protein by the host importin (IMP) α/ß1 heterodimer, and recently demonstrated its ability to bind directly to IMPα to cause conformational changes that prevent its function in nuclear import of key viral as well as host proteins. Cell culture experiments have shown robust antiviral action towards a whole range of viruses, including HIV-1, dengue, Zika and West Nile Virus, Venezuelan equine encephalitis virus, Chikungunya, pseudorabies virus, adenovirus, and SARS-CoV-2 (COVID-19). Close to 70 clinical trials are currently in progress worldwide for SARS-CoV-2. Although few of these studies have been completed, the results that are available, as well as those from observational/retrospective studies, indicate clinical benefit. Here we discuss the case for ivermectin as a host-directed broad-spectrum antiviral agent, including for SARS-CoV-2.


Asunto(s)
Antiparasitarios/farmacología , Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Ivermectina/farmacología , SARS-CoV-2/efectos de los fármacos , Antiparasitarios/uso terapéutico , Antivirales/uso terapéutico , Humanos , Ivermectina/uso terapéutico , alfa Carioferinas/antagonistas & inhibidores
8.
J Virol ; 94(18)2020 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-32641484

RESUMEN

Human adenoviruses (HAdV) are ubiquitous within the human population and comprise a significant burden of respiratory illnesses worldwide. Pediatric and immunocompromised individuals are at particular risk for developing severe disease; however, no approved antiviral therapies specific to HAdV exist. Ivermectin is an FDA-approved broad-spectrum antiparasitic drug that also exhibits antiviral properties against a diverse range of viruses. Its proposed function is inhibiting the classical protein nuclear import pathway mediated by importin-α (Imp-α) and -ß1 (Imp-ß1). Many viruses, including HAdV, rely on this host pathway for transport of viral proteins across the nuclear envelope. In this study, we show that ivermectin inhibits HAdV-C5 early gene transcription, early and late protein expression, genome replication, and production of infectious viral progeny. Similarly, ivermectin inhibits genome replication of HAdV-B3, a clinically important pathogen responsible for numerous recent outbreaks. Mechanistically, we show that ivermectin disrupts binding of the viral E1A protein to Imp-α without affecting the interaction between Imp-α and Imp-ß1. Our results further extend ivermectin's broad antiviral activity and provide a mechanistic underpinning for its mode of action as an inhibitor of cellular Imp-α/ß1-mediated nuclear import.IMPORTANCE Human adenoviruses (HAdVs) represent a ubiquitous and clinically important pathogen without an effective antiviral treatment. HAdV infections typically cause mild symptoms; however, individuals such as children, those with underlying conditions, and those with compromised immune systems can develop severe disseminated disease. Our results demonstrate that ivermectin, an FDA-approved antiparasitic agent, is effective at inhibiting replication of several HAdV types in vitro This is in agreement with the growing body of literature suggesting ivermectin has broad antiviral activity. This study expands our mechanistic knowledge of ivermectin by showing that ivermectin targets the ability of importin-α (Imp-α) to recognize nuclear localization sequences, without effecting the Imp-α/ß1 interaction. These data also exemplify the applicability of targeting host factors upon which viruses rely as a viable antiviral strategy.


Asunto(s)
Transporte Activo de Núcleo Celular/efectos de los fármacos , Adenovirus Humanos/efectos de los fármacos , Antiparasitarios/farmacología , Ivermectina/farmacología , Replicación Viral/efectos de los fármacos , alfa Carioferinas/genética , beta Carioferinas/genética , Células A549 , Transporte Activo de Núcleo Celular/genética , Adenovirus Humanos/genética , Adenovirus Humanos/metabolismo , Adenovirus Humanos/patogenicidad , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Núcleo Celular/virología , Citosol/efectos de los fármacos , Citosol/metabolismo , Citosol/virología , Fibroblastos/efectos de los fármacos , Fibroblastos/virología , Regulación de la Expresión Génica , Células HEK293 , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Humanos , Transducción de Señal , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/genética , Proteínas Virales/metabolismo , alfa Carioferinas/antagonistas & inhibidores , alfa Carioferinas/metabolismo , beta Carioferinas/metabolismo
9.
Antiviral Res ; 177: 104760, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32135219

RESUMEN

Infection by RNA viruses such as human immunodeficiency virus (HIV)-1, influenza, and dengue virus (DENV) represent a major burden for human health worldwide. Although RNA viruses replicate in the infected host cell cytoplasm, the nucleus is central to key stages of the infectious cycle of HIV-1 and influenza, and an important target of DENV nonstructural protein 5 (NS5) in limiting the host antiviral response. We previously identified the small molecule ivermectin as an inhibitor of HIV-1 integrase nuclear entry, subsequently showing ivermectin could inhibit DENV NS5 nuclear import, as well as limit infection by viruses such as HIV-1 and DENV. We show here that ivermectin's broad spectrum antiviral activity relates to its ability to target the host importin (IMP) α/ß1 nuclear transport proteins responsible for nuclear entry of cargoes such as integrase and NS5. We establish for the first time that ivermectin can dissociate the preformed IMPα/ß1 heterodimer, as well as prevent its formation, through binding to the IMPα armadillo (ARM) repeat domain to impact IMPα thermal stability and α-helicity. We show that ivermectin inhibits NS5-IMPα interaction in a cell context using quantitative bimolecular fluorescence complementation. Finally, we show for the first time that ivermectin can limit infection by the DENV-related West Nile virus at low (µM) concentrations. Since it is FDA approved for parasitic indications, ivermectin merits closer consideration as a broad spectrum antiviral of interest.


Asunto(s)
Transporte Activo de Núcleo Celular/efectos de los fármacos , Ivermectina/farmacología , alfa Carioferinas/antagonistas & inhibidores , beta Carioferinas/antagonistas & inhibidores , Animales , Línea Celular , Chlorocebus aethiops , Cricetinae , Infecciones por Flavivirus/tratamiento farmacológico , Riñón/citología , Unión Proteica , Células Vero , alfa Carioferinas/metabolismo , beta Carioferinas/metabolismo
10.
Virology ; 541: 124-135, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32056710

RESUMEN

Zika virus (ZIKV) nonstructural protein 5 (NS5) is a multifunctional protein possessing methyltransferase and RNA-dependent RNA polymerase activities. In the present study, we have carried out an extensive mutagenesis analysis to determine the importance of nuclear localization sequences (NLS) of NS5 in its nuclear accumulation and ZIKV replication. Deletion mutagenesis analysis demonstrated that the bipartite NLS consisting of importin ß1 (ßNLS) and importin α/ß-recognized NLS (α/ßNLS) is required for NS5 nuclear accumulation. Deletion of ßNLS, α/ßNLS, or both as well as R393A and R393N mutations severely impaired NS5 nuclear import and consequently conferred NS5 degradation. The R393A and R393N mutations also ablated viral RNA replication and virus production. Treatment of ZIKV-infected cells with importin α/ß-NS5 interaction inhibitors ivermectin or 4-HPR resulted in a rapid degradation of NS5 similar to the R393 A/N mutations. Collectively, these findings suggest that NS5 nuclear accumulation protects NS5 from cytoplasmic degradation and therefore is required for viral RNA replication.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas no Estructurales Virales/metabolismo , Replicación Viral , Virus Zika/fisiología , Transporte Activo de Núcleo Celular , Animales , Células Cultivadas , Citoplasma/metabolismo , Humanos , Ivermectina/farmacología , Señales de Localización Nuclear , alfa Carioferinas/antagonistas & inhibidores
11.
Sci Rep ; 9(1): 17620, 2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31772273

RESUMEN

Inactivation of the protein complex 'mechanistic target of rapamycin complex 1' (mTORC1) can increase the nuclear content of transcriptional regulators of metabolism and apoptosis. Previous studies established that nuclear import of signal transducer and activator of transcription-1 (STAT1) requires the mTORC1-associated adaptor karyopherin-α1 (KPNA1) when mTORC1 activity is reduced. However, the role of other mTORC1-interacting proteins in the complex, including 'protein kinase C delta' (PKCδ), have not been well characterized. In this study, we demonstrate that PKCδ, a STAT1 kinase, contains a functional 'target of rapamycin signaling' (TOS) motif that directs its interaction with mTORC1. Depletion of KPNA1 by RNAi prevented the nuclear import of PKCδ in cells exposed to the mTORC1 inhibitor rapamycin or amino acid restriction. Mutation of the TOS motif in PKCδ led to its loss of regulation by mTORC1 or karyopherin-α1, resulting in increased constitutive nuclear content. In cells expressing wild-type PKCδ, STAT1 activity and apoptosis were increased by rapamycin or interferon-ß. Those expressing the PKCδ TOS mutant exhibited increased STAT1 activity and apoptosis; further enhancement by rapamycin or interferon-ß, however, was lost. Therefore, the TOS motif in PKCδ is a novel structural mechanism by which mTORC1 prevents PKCδ and STAT1 nuclear import, and apoptosis.


Asunto(s)
Núcleo Celular/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteína Quinasa C-delta/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Secuencias de Aminoácidos , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Línea Celular , Humanos , Modelos Moleculares , Mutación Missense , Mutación Puntual , Conformación Proteica , Mapeo de Interacción de Proteínas , Proteína Quinasa C-delta/química , Proteína Quinasa C-delta/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteínas Recombinantes/metabolismo , Proteína Reguladora Asociada a mTOR/metabolismo , Factor de Transcripción STAT1/biosíntesis , Alineación de Secuencia , Sirolimus/farmacología , alfa Carioferinas/antagonistas & inhibidores , alfa Carioferinas/metabolismo
12.
Sci Rep ; 8(1): 6791, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29717156

RESUMEN

Under diabetic conditions, sodium-glucose cotransporter 2 (SGLT2) for glucose uptake in proximal tubules (PTs) increases, whereas NAD+-dependent protein deacetylase silent mating type information regulation 2 homolog 1 (Sirtuin-1; SIRT1) for PT survival decreases. Therefore, we hypothesized that increased glucose influx by SGLT2 reduces SIRT1 expression. To test this hypothesis, db/db mice with diabetes and high-glucose (HG)-cultured porcine PT LLC-PK1 cells in a two-chamber system were treated with the SGLT2 inhibitor canagliflozin. We also examined SIRT1 and SGLT2 expression in human kidney biopsies. In db/db mice, SGLT2 expression increased with concomitant decreases in SIRT1, but was inhibited by canagliflozin. For determination of the polarity of SGLT2 and SIRT1 expression, LLC-PK1 cells were seeded into Transwell chambers (pore size, 0.4 µm; Becton Dickinson, Oxford, UK). HG medium was added to either or to both of the upper and lower chambers, which corresponded to the apical and basolateral sides of the cells, respectively. In this system, the lower chamber with HG showed increased SGLT2 and decreased SIRT1 expression. Canagliflozin reversed HG-induced SIRT1 downregulation. Gene silencing and inhibitors for glucose transporter 2 (GLUT2) blocked HG-induced SGLT2 expression upregulation. Gene silencing for the hepatic nuclear factor-1α (HNF-1α), whose nuclear translocation was enhanced by HG, blocked HG-induced SGLT2 expression upregulation. Similarly, gene silencing for importin-α1, a chaperone protein bound to GLUT2, blocked HG-induced HNF-1α nuclear translocation and SGLT2 expression upregulation. In human kidney, SIRT1 immunostaining was negatively correlated with SGLT2 immunostaining. Thus, under diabetic conditions, SIRT1 expression in PTs was downregulated by an increase in SGLT2 expression, which was stimulated by basolateral HG through activation of the GLUT2/importin-α1/HNF-1α pathway.


Asunto(s)
Diabetes Mellitus Experimental/genética , Nefropatías Diabéticas/genética , Transportador de Glucosa de Tipo 2/genética , Glucosa/farmacología , Sirtuina 1/genética , Transportador 2 de Sodio-Glucosa/genética , Animales , Canagliflozina/farmacología , Línea Celular , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Cámaras de Difusión de Cultivos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Regulación de la Expresión Génica , Transportador de Glucosa de Tipo 2/antagonistas & inhibidores , Transportador de Glucosa de Tipo 2/metabolismo , Factor Nuclear 1-alfa del Hepatocito/antagonistas & inhibidores , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Humanos , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Sirtuina 1/metabolismo , Transportador 2 de Sodio-Glucosa/metabolismo , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Porcinos , alfa Carioferinas/antagonistas & inhibidores , alfa Carioferinas/genética , alfa Carioferinas/metabolismo
13.
Oncol Rep ; 40(1): 179-194, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29781035

RESUMEN

Oral squamous cell carcinoma (OSCC), one of the 10 most common types of neoplasms in the US, constitutes ~90% of all cases of oral malignancies. Chemoresistance and metastasis are difficult to avoid during the course of treatment, leading to a poor prognosis and a high mortality rate for patients with OSCC. Autophagy, a critical conserved cellular process, has been reported to be highly associated with the regulation of chemoresistance and metastasis of cancer cells. The present study investigated the role of karyopherin α2 (KPNA2), a member of the importin α family, which may serve an important role in p53 nucleocytoplasmic transport in the process of OSCC autophagy. In the CAL­27, SCC­15 and Tca8113 OSCC cell lines, we observed that the downregulation of KPNA2 suppressed cell migration and cisplatin resistance, using wound­healing, Transwell and CCK­8 assays. Additionally, the results of western blot analysis and transmission electron microscopy (TEM) analysis indicated that the knockdown of KPNA2 inhibited autophagy. We confirmed that the inhibition of autophagy with anti­autophagy agents decreased the migration and cisplatin resistance of OSCC cells. We hypothesized that the suppression of cell migration and cisplatin resistance induced by KPNA2 knockdown may be associated with the inhibition of autophagy. To identify the underlying mechanism, further experiments determined that KPNA2 affects the level of autophagy via regulating the p53 nuclear import. Thus, the present study demonstrated that the function of KPNA2 in the process of autophagy may be p53­dependent, and by regulating the translocation of p53, KPNA2 can support autophagy to promote the chemoresistance and metastasis of OSCC cells.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Neoplasias de la Boca/tratamiento farmacológico , Proteína p53 Supresora de Tumor/genética , alfa Carioferinas/genética , Transporte Activo de Núcleo Celular/genética , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Autofagia/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Linaje de la Célula/genética , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Transporte de Proteínas/genética , alfa Carioferinas/antagonistas & inhibidores
14.
Cell ; 173(4): 958-971.e17, 2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29628143

RESUMEN

Defects in nucleocytoplasmic transport have been identified as a key pathogenic event in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) mediated by a GGGGCC hexanucleotide repeat expansion in C9ORF72, the most common genetic cause of ALS/FTD. Furthermore, nucleocytoplasmic transport disruption has also been implicated in other neurodegenerative diseases with protein aggregation, suggesting a shared mechanism by which protein stress disrupts nucleocytoplasmic transport. Here, we show that cellular stress disrupts nucleocytoplasmic transport by localizing critical nucleocytoplasmic transport factors into stress granules, RNA/protein complexes that play a crucial role in ALS pathogenesis. Importantly, inhibiting stress granule assembly, such as by knocking down Ataxin-2, suppresses nucleocytoplasmic transport defects as well as neurodegeneration in C9ORF72-mediated ALS/FTD. Our findings identify a link between stress granule assembly and nucleocytoplasmic transport, two fundamental cellular processes implicated in the pathogenesis of C9ORF72-mediated ALS/FTD and other neurodegenerative diseases.


Asunto(s)
Transporte Activo de Núcleo Celular/fisiología , Esclerosis Amiotrófica Lateral/patología , Ataxina-2/metabolismo , Proteína C9orf72/genética , Demencia Frontotemporal/patología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Anciano , Esclerosis Amiotrófica Lateral/metabolismo , Arsenitos/toxicidad , Ataxina-2/antagonistas & inhibidores , Ataxina-2/genética , Proteína C9orf72/metabolismo , Expansión de las Repeticiones de ADN/genética , Femenino , Demencia Frontotemporal/metabolismo , Células HEK293 , Humanos , Masculino , Glicoproteínas de Membrana/metabolismo , Persona de Mediana Edad , Proteínas de Complejo Poro Nuclear/metabolismo , Estrés Oxidativo/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Compuestos de Sodio/toxicidad , alfa Carioferinas/antagonistas & inhibidores , alfa Carioferinas/genética , alfa Carioferinas/metabolismo , beta Carioferinas/antagonistas & inhibidores , beta Carioferinas/genética , beta Carioferinas/metabolismo , Proteína de Unión al GTP ran/antagonistas & inhibidores , Proteína de Unión al GTP ran/genética , Proteína de Unión al GTP ran/metabolismo
15.
Sci Rep ; 7(1): 17705, 2017 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-29255256

RESUMEN

Therapeutics are currently unavailable for Venezuelan equine encephalitis virus (VEEV), which elicits flu-like symptoms and encephalitis in humans, with an estimated 14% of cases resulting in neurological disease. Here we identify anti-VEEV agents using in silico structure-based-drug-design (SBDD) for the first time, characterising inhibitors that block recognition of VEEV capsid protein (C) by the host importin (IMP) α/ß1 nuclear transport proteins. From an initial screen of 1.5 million compounds, followed by in silico refinement and screening for biological activity in vitro, we identified 21 hit compounds which inhibited IMPα/ß1:C binding with IC50s as low as 5 µM. Four compounds were found to inhibit nuclear import of C in transfected cells, with one able to reduce VEEV replication at µM concentration, concomitant with reduced C nuclear accumulation in infected cells. Further, this compound was inactive against a mutant VEEV that lacks high affinity IMPα/ß1:C interaction, supporting the mode of its antiviral action to be through inhibiting C nuclear localization. This successful application of SBDD paves the way for lead optimization for VEEV antivirals, and is an exciting prospect to identify inhibitors for the many other viral pathogens of significance that require IMPα/ß1 in their infectious cycle.


Asunto(s)
Proteínas de la Cápside/efectos de los fármacos , Descubrimiento de Drogas/métodos , Virus de la Encefalitis Equina Venezolana/efectos de los fármacos , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Antivirales/farmacología , Cápside , Proteínas de la Cápside/metabolismo , Núcleo Celular/metabolismo , Chlorocebus aethiops , Simulación por Computador , Diseño de Fármacos , Virus de la Encefalitis Equina Venezolana/patogenicidad , Humanos , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Células Vero , Replicación Viral/efectos de los fármacos , alfa Carioferinas/antagonistas & inhibidores , alfa Carioferinas/metabolismo , beta Carioferinas/antagonistas & inhibidores , beta Carioferinas/metabolismo
16.
Endocr J ; 64(10): 963-975, 2017 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-28835592

RESUMEN

Insulin-like growth factor-binding protein (IGFBP)-5 is a secreted protein that binds to IGFs and modulates IGF actions, as well as regulates cell proliferation, migration, and apoptosis independent of IGF. Proper cellular localization is critical for the effective function of most signaling molecules. In previous studies, we have shown that the nuclear IGFBP-5 comes from ER-cytosol retro-translocation. In this study, we further investigated the pathway mediating IGFBP-5 nuclear import after it retro-translocation. Importin-α5 was identified as an IGFBP-5-interacting protein with a yeast two-hybrid system, and its interaction with IGFBP-5 was further confirmed by GST pull down and co-immunoprecipitation. Binding affinity of IGFBP-5 and importins were determined by surface plasmon resonance (IGFBP-5/importin-ß: KD=2.44e-7, IGFBP-5/importin-α5: KD=3.4e-7). Blocking the importin-α5/importin-ß nuclear import pathway using SiRNA or dominant negative impotin-ß dramatically inhibited IGFBP-5-EGFP nuclear import, though importin-α5 overexpress does not affect IGFBP-5 nuclear import. Furthermore, nuclear IGFBP-5 was quantified using luciferase report assay. When deleted the IGFBP-5 nuclear localization sequence (NLS), IGFBP-5ΔNLS loss the ability to translocate into the nucleus and accumulation of IGFBP-5ΔNLS was visualized in the cytosol. Altogether, our findings provide a substantially evidence showed that the IGFBP-5 nuclear import is mediated by importin-α/importin-ß complex, and NLS is critical domain in IGFBP-5 nuclear translocation.


Asunto(s)
Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , alfa Carioferinas/metabolismo , beta Carioferinas/metabolismo , Transporte Activo de Núcleo Celular , Eliminación de Gen , Genes Reporteros , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células HeLa , Humanos , Inmunoprecipitación , Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/química , Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Cinética , Microscopía Confocal , Microscopía Fluorescente , Mutación , Señales de Localización Nuclear/antagonistas & inhibidores , Señales de Localización Nuclear/genética , Señales de Localización Nuclear/metabolismo , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Multimerización de Proteína , Interferencia de ARN , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Resonancia por Plasmón de Superficie , Técnicas del Sistema de Dos Híbridos , alfa Carioferinas/antagonistas & inhibidores , alfa Carioferinas/química , alfa Carioferinas/genética , beta Carioferinas/antagonistas & inhibidores , beta Carioferinas/química , beta Carioferinas/genética
17.
PLoS One ; 12(6): e0179468, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28628637

RESUMEN

The rising tide of sepsis, a leading cause of death in the US and globally, is not adequately controlled by current antimicrobial therapies and supportive measures, thereby requiring new adjunctive treatments. Severe microvascular injury and multiple organ failure in sepsis are attributed to a "genomic storm" resulting from changes in microbial and host genomes encoding virulence factors and endogenous inflammatory mediators, respectively. This storm is mediated by stress-responsive transcription factors that are ferried to the nucleus by nuclear transport shuttles importins/karyopherins. We studied the impact of simultaneously targeting two of these shuttles, importin alpha 5 (Imp α5) and importin beta 1 (Imp ß1), with a cell-penetrating Nuclear Transport Modifier (NTM) in a mouse model of polymicrobial sepsis. NTM reduced nuclear import of stress-responsive transcription factors nuclear factor kappa B, signal transducer and activator of transcription 1 alpha, and activator protein 1 in liver, which was also protected from sepsis-associated metabolic changes. Strikingly, NTM without antimicrobial therapy improved bacterial clearance in blood, spleen, and lungs, wherein a 700-fold reduction in bacterial burden was achieved while production of proinflammatory cytokines and chemokines in blood plasma was suppressed. Furthermore, NTM significantly improved thrombocytopenia, a prominent sign of microvascular injury in sepsis, inhibited neutrophil infiltration in the liver, decreased L-selectin, and normalized plasma levels of E-selectin and P-selectin, indicating reduced microvascular injury. Importantly, NTM combined with antimicrobial therapy extended the median time to death from 42 to 83 hours and increased survival from 30% to 55% (p = 0.022) as compared to antimicrobial therapy alone. This study documents the fundamental role of nuclear signaling mediated by Imp α5 and Imp ß1 in the mechanism of polymicrobial sepsis and highlights the potential for targeting nuclear transport as an adjunctive therapy in sepsis management.


Asunto(s)
Transporte Activo de Núcleo Celular/efectos de los fármacos , Péptidos de Penetración Celular/farmacología , Proteínas Nucleares/metabolismo , Sepsis/patología , alfa Carioferinas/metabolismo , Animales , Antiinfecciosos/uso terapéutico , Péptidos de Penetración Celular/síntesis química , Péptidos de Penetración Celular/química , Quimiocinas/sangre , Citocinas/sangre , Modelos Animales de Enfermedad , Femenino , Hígado/inmunología , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Neutrófilos/citología , Neutrófilos/inmunología , Proteínas Nucleares/antagonistas & inhibidores , Selectinas/sangre , Sepsis/tratamiento farmacológico , Sepsis/microbiología , Sepsis/mortalidad , Tasa de Supervivencia , Trombocitopenia/patología , Factor de Transcripción ReIA/metabolismo , alfa Carioferinas/antagonistas & inhibidores , beta Carioferinas
18.
J Biol Chem ; 292(24): 10262-10274, 2017 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-28455446

RESUMEN

Enterovirus 71 (EV71) has emerged as one of the most important enteroviruses since the eradication of poliovirus, and it causes severe neurological symptoms for which no effective antiviral drugs are available. Type I interferons (IFN) α/ß have been used clinically as antiviral therapy as the first line of defense against virus infections successfully for decades. However, treatment with type I interferons has not been effective in patients with EV71 infection. In this study, we found that in cells pretreated with IFN-ß, EV71 infection could still lead to a cytopathic effect, and the viral replication was not affected. The mechanism by which EV71 antagonizes interferon signaling, however, has been controversial. Our study indicated that EV71 infection did not inhibit phosphorylation of STAT1/2 induced by IFN-ß stimulation, but p-STAT1/2 transport into the nucleus was significantly blocked. We showed that EV71 infection reduced the formation of STAT/karyopherin-α1 (KPNA1) complex upon interferon stimulation and that the virus down-regulated the expression of KPNA1, a nuclear localization signal receptor for p-STAT1. Using specific caspase inhibitors and siRNA for caspase-3, we demonstrated that EV71 infection induced degradation of cellular KPNA1 in a caspase-3-dependent manner, which led to decreased induction of interferon-inducible genes and IFN response. Viral 2A and 3C proteases did not degrade KPNA1, inhibit the activity of ISRE or suppress the transcription of interferon-inducible genes induced by IFN-ß. Our study demonstrates a novel mechanism by which antiviral signaling is suppressed through degradation of KPNA1 by activated caspase-3 induced in an enteroviral infection.


Asunto(s)
Caspasa 3/metabolismo , Enterocitos/virología , Enterovirus Humano A/fisiología , Interferón beta/metabolismo , Janus Quinasa 1/metabolismo , Transducción de Señal , alfa Carioferinas/antagonistas & inhibidores , Transporte Activo de Núcleo Celular , Animales , Caspasa 3/química , Caspasa 3/genética , Chlorocebus aethiops , Enterocitos/inmunología , Enterocitos/metabolismo , Enterovirus Humano A/crecimiento & desarrollo , Células HT29 , Células HeLa , Humanos , Interferón beta/genética , Janus Quinasa 1/genética , Fosforilación , Procesamiento Proteico-Postraduccional , Proteolisis , Interferencia de ARN , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo , Células Vero , Replicación Viral , alfa Carioferinas/genética , alfa Carioferinas/metabolismo
19.
Oncogene ; 36(20): 2868-2878, 2017 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-27941876

RESUMEN

Prostate cancer (PCa) is a common cancer in men. Although current treatments effectively palliate symptoms and prolong life, the metastatic PCa remains incurable. It is important to find biomarkers and targets to improve metastatic PCa diagnosis and treatment. Here we report a novel correlation between karyopherin α4 (KPNA4) and PCa pathological stages. KPNA4 mediates the cytoplasm-to-nucleus translocation of transcription factors, including nuclear factor kappa B, although its role in PCa was largely unknown. We find that knockdown of KPNA4 reduces cell migration in multiple PCa cell lines, suggesting a role of KPNA4 in PCa progression. Indeed, stable knockdown of KPNA4 significantly reduces PCa invasion and distant metastasis in mouse models. Functionally, KPNA4 alters tumor microenvironment in terms of macrophage polarization and osteoclastogenesis by modulating tumor necrosis factor (TNF)-α and -ß. Further, KPNA4 is proved as a direct target of miR-708, a tumor-suppressive microRNA. We disclose the role of miR-708-KPNA4-TNF axes in PCa metastasis and KPNA4's potential as a novel biomarker for PCa metastasis.


Asunto(s)
Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , alfa Carioferinas/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Progresión de la Enfermedad , Expresión Génica , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Humanos , Linfotoxina-alfa/metabolismo , Masculino , Ratones , MicroARNs/genética , Modelos Biológicos , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias de la Próstata/genética , Interferencia de ARN , Factor de Necrosis Tumoral alfa/metabolismo , alfa Carioferinas/genética , alfa Carioferinas/metabolismo
20.
Mol Med Rep ; 15(2): 551-558, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28000846

RESUMEN

Osteoarthritis (OA) is a common chronic joint disease, the etiology of which is complex. Disturbance to proinflammatory and anti­inflammatory signaling pathways is a major cause of OA. MicroRNAs (miRNAs/miR) are a group of endogenous, short, non­coding RNAs, the expression profile of which is disturbed in the cartilage of patients with OA. To determine the function of miRNAs during the progression of OA, the present study detected the expression levels of nine candidate miRNAs in cartilage samples from 33 patients with OA. The results demonstrated that miR­26a, miR­26b, miR­138 and miR­140 were downregulated in patients with OA. As predicted by a bioinformatics analysis and confirmed by luciferase assay and western blotting, the present study revealed that miR­26a and miR­26b are able to suppress karyopherin subunit alpha 3 (KPNA3) expression by targeting its 3'­untranslated region. Since KPNA3 is an important mediator that modulates nuclear factor (NF)­κB p65 translocation, the present study examined the impact of miR­26a and miR­26b on NF­κB signaling. The results indicated that transfection of cells with a miR­26a or miR­26b inhibitor may promote NF­κB p65 translocation from the cytoplasm to the nucleus via the upregulation of KPNA3. Furthermore, the expression levels of matrix metalloproteinase­3, ­9, ­13 and cyclooxygenase­2 were upregulated following transfection with a miR­26a or miR­26b inhibitor. These results indicate that downregulation of miR­26a and miR­26b may contribute to the pathogenesis of OA via promotion of the NF­κB signaling pathway. The present study sheds light on the pathogenesis of OA and may provide a target for the development of therapeutic methods for the treatment of OA.


Asunto(s)
MicroARNs/metabolismo , Osteoartritis/patología , Factor de Transcripción ReIA/metabolismo , alfa Carioferinas/metabolismo , Regiones no Traducidas 3' , Adulto , Anciano , Antagomirs/metabolismo , Cartílago/metabolismo , Células Cultivadas , Condrocitos/citología , Condrocitos/metabolismo , Ciclooxigenasa 2/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Femenino , Humanos , Interleucina-1beta/farmacología , Masculino , Metaloproteinasas de la Matriz/metabolismo , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Persona de Mediana Edad , Osteoartritis/genética , Osteoartritis/metabolismo , Transducción de Señal , Regulación hacia Arriba/efectos de los fármacos , alfa Carioferinas/antagonistas & inhibidores , alfa Carioferinas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...