Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell ; 186(16): 3427-3442.e22, 2023 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-37421949

RESUMEN

SARS-CoV-2 is associated with broad tissue tropism, a characteristic often determined by the availability of entry receptors on host cells. Here, we show that TMEM106B, a lysosomal transmembrane protein, can serve as an alternative receptor for SARS-CoV-2 entry into angiotensin-converting enzyme 2 (ACE2)-negative cells. Spike substitution E484D increased TMEM106B binding, thereby enhancing TMEM106B-mediated entry. TMEM106B-specific monoclonal antibodies blocked SARS-CoV-2 infection, demonstrating a role of TMEM106B in viral entry. Using X-ray crystallography, cryogenic electron microscopy (cryo-EM), and hydrogen-deuterium exchange mass spectrometry (HDX-MS), we show that the luminal domain (LD) of TMEM106B engages the receptor-binding motif of SARS-CoV-2 spike. Finally, we show that TMEM106B promotes spike-mediated syncytium formation, suggesting a role of TMEM106B in viral fusion. Together, our findings identify an ACE2-independent SARS-CoV-2 infection mechanism that involves cooperative interactions with the receptors heparan sulfate and TMEM106B.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , Receptores Virales/metabolismo , Internalización del Virus , Unión Proteica , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo
2.
mBio ; 13(4): e0137622, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-35913162

RESUMEN

The continuous emergence of new variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) urges better understanding of the functional motifs in the spike (S) protein and their tolerance to mutations. Here, we focused on the S2' motif, which, during virus entry, requires cleavage by a host cell protease to release the fusion peptide. Though belonging to an immunogenic region, the SARS-CoV-2 S2' motif (811-KPSKR-815) has shown hardly any variation, with its three basic (K/R) residues being >99.99% conserved thus far. By creating a series of mutant pseudoviruses bearing the spikes of Wuhan-Hu-1, its G614 mutant or the Delta and Omicron variants, we show that residue K814 (preceding the scissile R815) is dispensable for TMPRSS2 yet favored by the alternative TMPRSS13 protease. Activation by TMPRSS13 was drastically reduced when the SARS-CoV-2 S2' motif was swapped with that of the low pathogenic 229E coronavirus (685-RVAGR-689), and also, the reverse effect was seen. This swap had no impact on recognition by TMPRSS2. In the Middle East respiratory syndrome coronavirus (MERS-CoV) spike, introducing a dibasic scissile motif was easily accepted by TMPRSS13 but less so by TMPRSS2, confirming that TMPRSS13 favors a sequence rich in K/R residues. Pseudovirus entry experiments in Calu-3 cells confirmed that the S2' mutations have minor impact on TMPRSS2. Our findings are the first to demonstrate which S2' residues are important for SARS-CoV-2 spike activation by these two airway proteases, with TMPRSS2 being more tolerant to variation than TMPRSS13. This preemptive insight will help to estimate the impact of S2' motif changes as they appear in new SARS-CoV-2 variants. IMPORTANCE Since its introduction in humans, SARS-CoV-2 is evolving with frequent appearance of new variants. The surveillance would benefit from proactive characterization of the functional motifs in the spike (S) protein, the most variable viral factor. This is linked to immune evasion but also influences spike functioning. Remarkably, though located in a strongly immunogenic region, the S2' cleavage motif has, thus far, remained highly conserved. This suggests that its sequence is critical for spike activation by airway proteases. To investigate this, we assessed how pseudovirus entry is affected by changes in the S2' motif. We demonstrate that TMPRSS2 readily accepts variations in this motif, whereas the alternative TMPRSS13 protease is more fastidious. The Wuhan-Hu-1, G614, Delta and Omicron spikes showed no difference in this regard. Being the first in its kind, our study will help to assess the impact of S2' variations as soon as they are detected during variant surveillance.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Proteínas de la Membrana/genética , Mutación , Péptido Hidrolasas/genética , SARS-CoV-2/genética , Serina Endopeptidasas/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus
3.
Pharmaceuticals (Basel) ; 14(10)2021 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-34681276

RESUMEN

We used classical linear and microwave-assisted synthesis methods to prepare novel N-substituted, benzimidazole-derived acrylonitriles with antiproliferative activity against several cancer cells in vitro. The most potent systems showed pronounced activity against all tested hematological cancer cell lines, with favorable selectivity towards normal cells. The selection of lead compounds was also tested in vitro for tubulin polymerization inhibition as a possible mechanism of biological action. A combination of docking and molecular dynamics simulations confirmed the suitability of the employed organic skeleton for the design of antitumor drugs and demonstrated that their biological activity relies on binding to the colchicine binding site in tubulin. In addition, it also underlined that higher tubulin affinities are linked with (i) bulkier alkyl and aryl moieties on the benzimidazole nitrogen and (ii) electron-donating substituents on the phenyl group that allow deeper entrance into the hydrophobic pocket within the tubulin's ß-subunit, consisting of Leu255, Leu248, Met259, Ala354, and Ile378 residues.

4.
Nat Microbiol ; 6(10): 1219-1232, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34471255

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic has claimed millions of lives and caused a global economic crisis. No effective antiviral drugs are currently available to treat infections of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The medical need imposed by the pandemic has spurred unprecedented research efforts to study coronavirus biology. Every virus depends on cellular host factors and pathways for successful replication. These proviral host factors represent attractive targets for antiviral therapy as they are genetically more stable than viral targets and may be shared among related viruses. The application of various 'omics' technologies has led to the rapid discovery of proviral host factors that are required for the completion of the SARS-CoV-2 life cycle. In this Review, we summarize insights into the proviral host factors that are required for SARS-CoV-2 infection that were mainly obtained using functional genetic and interactome screens. We discuss cellular processes that are important for the SARS-CoV-2 life cycle, as well as parallels with non-coronaviruses. Finally, we highlight host factors that could be targeted by clinically approved molecules and molecules in clinical trials as potential antiviral therapies for COVID-19.


Asunto(s)
COVID-19/metabolismo , SARS-CoV-2/fisiología , Antivirales/farmacología , Antivirales/uso terapéutico , COVID-19/virología , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Péptido Hidrolasas/metabolismo , ARN Viral/metabolismo , Receptores Virales/metabolismo , SARS-CoV-2/efectos de los fármacos , Proteínas Virales/metabolismo , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
5.
Nat Genet ; 53(4): 435-444, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33686287

RESUMEN

The ongoing COVID-19 pandemic has caused a global economic and health crisis. To identify host factors essential for coronavirus infection, we performed genome-wide functional genetic screens with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and human coronavirus 229E. These screens uncovered virus-specific as well as shared host factors, including TMEM41B and PI3K type 3. We discovered that SARS-CoV-2 requires the lysosomal protein TMEM106B to infect human cell lines and primary lung cells. TMEM106B overexpression enhanced SARS-CoV-2 infection as well as pseudovirus infection, suggesting a role in viral entry. Furthermore, single-cell RNA-sequencing of airway cells from patients with COVID-19 demonstrated that TMEM106B expression correlates with SARS-CoV-2 infection. The present study uncovered a collection of coronavirus host factors that may be exploited to develop drugs against SARS-CoV-2 infection or future zoonotic coronavirus outbreaks.


Asunto(s)
COVID-19/genética , Sistemas CRISPR-Cas , Genoma Humano/genética , Estudio de Asociación del Genoma Completo/métodos , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Líquido del Lavado Bronquioalveolar/citología , COVID-19/epidemiología , COVID-19/virología , Línea Celular Tumoral , Células Cultivadas , Coronavirus Humano 229E/genética , Epidemias , Células Epiteliales/virología , Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Provirus/fisiología , SARS-CoV-2/fisiología , Internalización del Virus
6.
J Gen Virol ; 100(4): 583-601, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30762518

RESUMEN

The possible resistance of influenza virus against existing antiviral drugs calls for new therapeutic concepts. One appealing strategy is to inhibit virus entry, in particular at the stage of internalization. This requires a better understanding of virus-host interactions during the entry process, including the role of receptor tyrosine kinases (RTKs). To search for cellular targets, we evaluated a panel of 276 protein kinase inhibitors in a multicycle antiviral assay in Madin-Darby canine kidney cells. The RTK inhibitor Ki8751 displayed robust anti-influenza A and B virus activity and was selected for mechanistic investigations. Ki8751 efficiently disrupted the endocytic process of influenza virus in different cell lines carrying platelet-derived growth factor receptor ß (PDGFRß), an RTK that is known to act at GM3 ganglioside-positive lipid rafts. The more efficient virus entry in CHO-K1 cells compared to the wild-type ancestor (CHO-wt) cells indicated a positive effect of GM3, which is abundant in CHO-K1 but not in CHO-wt cells. Entering virus localized to GM3-positive lipid rafts and the PDGFRß-containing endosomal compartment. PDGFRß/GM3-dependent virus internalization involved PDGFRß phosphorylation, which was potently inhibited by Ki8751, and desialylation of activated PDGFRß by the viral neuraminidase. Virus uptake coincided with strong activation of the Raf/MEK/Erk cascade, but not of PI3K/Akt or phospholipase C-γ. We conclude that influenza virus efficiently hijacks the GM3-enhanced PDGFRß signalling pathway for cell penetration, providing an opportunity for host cell-targeting antiviral intervention.


Asunto(s)
Gangliósido G(M3)/metabolismo , Gripe Humana/metabolismo , Gripe Humana/virología , Infecciones por Orthomyxoviridae/metabolismo , Orthomyxoviridae/patogenicidad , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/fisiología , Animales , Células CHO , Línea Celular , Cricetulus , Perros , Células HEK293 , Humanos , Gripe Humana/tratamiento farmacológico , Células de Riñón Canino Madin Darby , Orthomyxoviridae/efectos de los fármacos , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Compuestos de Fenilurea/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Quinolinas/farmacología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal/efectos de los fármacos , Internalización del Virus/efectos de los fármacos
7.
J Infect ; 77(2): 119-130, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29752963

RESUMEN

OBJECTIVES: The opportunistic pathogen Staphylococcus epidermidis is progressively involved in device-related infections. Since these infections involve biofilm formation, antibiotics are not effective. Conversely, a vaccine can be advantageous to prevent these infections. In view of vaccine development, predicted surface proteins were evaluated on their potential as a vaccine target. METHODS: Immunoglobulins directed against S. epidermidis surface proteins SesB, M, O, Q and R were used to firstly affirm their surface location. Further, inhibitory effects of these IgGs on biofilm formation were determined in vitro on polystyrene and polyurethane surfaces and in vivo using a subcutaneous catheter mouse model. We also examined the opsonophagocytotic capacity of these IgGs. RESULTS: Surface localization of the five Ses proteins was demonstrated both for planktonic and sessile cells, though to a variable extent. Ses-specific IgGs added to planktonic cells had a variable inhibitory effect on cell adhesion to polystyrene, while only anti-SesO IgGs decreased cell attachment to polyurethane catheters. Although phagocytic killing was only obtained after opsonization with SesB-specific IgGs, a significant reduction of in vivo formed biofilms was observed after administration of SesB-, SesM- and SesO-specific IgGs. CONCLUSIONS: Regardless of their characterization or function, S. epidermidis surface proteins can be adequate targets for vaccine development aiming the prevention of device-related infections caused by invasive S. epidermidis strains.


Asunto(s)
Proteínas Bacterianas/inmunología , Vacunas Bacterianas/inmunología , Proteínas de la Membrana/inmunología , Infecciones Estafilocócicas/prevención & control , Staphylococcus epidermidis/metabolismo , Animales , Especificidad de Anticuerpos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biopelículas , Regulación Bacteriana de la Expresión Génica/fisiología , Células HL-60 , Humanos , Inmunoglobulina G/inmunología , Conejos , Infecciones Estafilocócicas/microbiología , Staphylococcus epidermidis/inmunología
8.
Int J Food Microbiol ; 266: 190-199, 2018 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-29227906

RESUMEN

Postharvest losses of fruit and vegetables can reach up to 30%, the main cause being microbial decay. For apple fruit, mostly fungal pathogens, such as Penicillium expansum, Colletotrichum spp., Neofabraea spp. and Botrytis cinerea, are important. As such losses are unsustainable in many ways, it is necessary that research is conducted to prevent them. Generally, for plants and fruit grown under non-sterile field conditions, disinfection is carried out prior to the start of a phytopathological experiment. The motivation for this practice is the removal of background contamination so that it will not affect the experimental outcome and its interpretation. In literature, a plethora of disinfection methods exists, differing in disinfectant, strength and duration. The following two disinfectants are commonly used: sodium hypochlorite (NaOCl) and ethanol. This article presents a targeted investigation into the effects of these two disinfectants on apple fruit surface and physiology. The results clearly demonstrate that both were affected by both disinfectants. NaOCl caused oxidative damage to the apple's wax layer, causing it to crack. Ethanol affected a redistribution of the wax on the fruit surface and altered the wax composition and/or metabolism. Both NaOCl and ethanol treatment resulted in an increased respiration rate. Therefore, apple and possibly other fruit should not be disinfected in phytopathological studies. A negative control, as is typically used, is not solving this issue, as we clearly demonstrate that the living tissue shows metabolic effects following disinfection, and hence the study objects are changed, hampering a clear interpretation of the experimental outcomes. Moreover, fungal inoculation during experiments is typically taking place at rather large levels in wounded tissue (as infection success is the exception), outnumbering the variable levels of background population, if present.


Asunto(s)
Etanol/farmacología , Microbiología de Alimentos , Conservación de Alimentos/métodos , Frutas/efectos de los fármacos , Malus/efectos de los fármacos , Hipoclorito de Sodio/farmacología , Desinfectantes/farmacología , Microbiología de Alimentos/tendencias , Frutas/microbiología , Malus/microbiología
9.
Cell Tissue Res ; 370(3): 403-416, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28963588

RESUMEN

With most research on interstitial cells (IC) in the bladder being conducted on animal models, it remains unclear whether all structural and functional data on IC from animal models can be translated to the human context. This prompted us to compare the structural and immunohistochemical properties of IC in bladders from mouse, rat and human. Tissue samples were obtained from the bladder dome and subsequently processed for immunohistochemistry and electron microscopy. The ultrastructural properties of IC were compared by means of electron microscopy and IC were additionally characterized with single/double immunohistochemistry/immunofluorescence. Our results reveal a similar organization of the IC network in the upper lamina propria (ULP), the deep lamina propria (DLP) and the detrusor muscle in human, rat and mouse bladders. Furthermore, despite several similarities in IC phenotypes, we also found several obvious inter-species differences in IC, especially in the ULP. Most remarkably in this respect, ULP IC in human bladder predominantly displayed a myoid phenotype with abundant presence of contractile micro-filaments, while those in rat and mouse bladders showed a fibroblast phenotype. In conclusion, the organization of ULP IC, DLP IC and detrusor IC is comparable in human, rat and mouse bladders, although several obvious inter-species differences in IC phenotypes were found. The present data show that translating research data on IC in laboratory animals to the human setting should be carried out with caution.


Asunto(s)
Células Intersticiales de Cajal/ultraestructura , Membrana Mucosa/ultraestructura , Miocitos del Músculo Liso/ultraestructura , Vejiga Urinaria/anatomía & histología , Animales , Femenino , Humanos , Inmunohistoquímica , Masculino , Ratones , Microscopía Electrónica , Ratas , Ratas Sprague-Dawley
10.
Eur J Med Chem ; 132: 219-235, 2017 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-28365318

RESUMEN

5-(2-(4-Methoxyphenyl)ethyl)-2-amino-3-methylcarboxylate thiophene (TR560) is the prototype drug of a recently discovered novel class of tumor-selective compounds that preferentially inhibit the proliferation of specific tumor cell types (e.g. leukemia/lymphoma). Here, we further increased tumor selectivity by simplification of the molecule through replacing the 4-methoxyphenyl moiety by an alkyl chain. Several 2-amino-3-methylcarboxylate thiophene derivatives containing at C-5 an alkyl group consisting of at least 6 (hexyl) to 9 (nonyl) carbon units showed pronounced anti-proliferative activity in the mid-nanomolar range with 500- to 1000-fold tumor cell selectivity. The compounds preferentially inhibited the proliferation of T-lymphoma CEM and Molt/4, prostate PC-3, kidney Caki-1 and hepatoma Huh-7 tumor cells, but were virtually inactive against other tumor cell lines including B-lymphoma Raji and cervix carcinoma HeLa cells. The novel prototype drug 3j (containing a 5-heptyl chain) elicited a cytotoxic, rather than cytostatic activity, already after 4 h of exposure. The unusual tumor selectivity could not be explained by a differential uptake (or efflux) of the drug by sensitive versus resistant tumor cells. Exposure of a fluorescent derivative of 3j revealed pronounced uptake of the drug in the cytoplasm, no visible appearance in the nucleus, and a predominant localization in the endoplasmic reticulum. These observations may be helpful to narrow down the intracellular localization and identification of the molecular target of the 5-substituted thiophene derivatives.


Asunto(s)
Antineoplásicos/farmacología , Tiofenos/síntesis química , Alquilación , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Humanos , Relación Estructura-Actividad , Tiofenos/farmacocinética , Tiofenos/farmacología , Distribución Tisular
11.
J Cell Mol Med ; 21(6): 1206-1216, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27997763

RESUMEN

The mast/stem cell growth factor receptor KIT has long been assumed to be a specific marker for interstitial cells of Cajal (ICC) in the bladder, with possible druggable perspectives. However, several authors have challenged the presence of KIT+ ICC in recent years. The aim of this study was therefore to attempt to clarify the conflicting reports on KIT expression in the bladder of human beings, rat, mouse and guinea pig and to elucidate the possible role of antibody-related issues and interspecies differences in this matter. Fresh samples were obtained from human, rat, mouse and guinea pig cystectomies and processed for single/double immunohistochemistry/immunofluorescence. Specific antibodies against KIT, mast cell tryptase (MCT), anoctamin-1 (ANO1) and vimentin were used to characterize the cell types expressing KIT. Gut (jejunum) tissue was used as an external antibody control. Our results revealed KIT expression on mast cells but not on ICC in human, rat, mouse and guinea pig bladder. Parallel immunohistochemistry showed KIT expression on ICC in human, rat, mouse and guinea pig gut, which confirmed the selectivity of the KIT antibody clones. In conclusion, we have shown that KIT+ cells in human, rat, mouse and guinea pig bladder are mast cells and not ICC. The present report is important as it opposes the idea that KIT+ ICC are present in bladder. In this perspective, functional concepts of KIT+ ICC being involved in sensory and/or motor aspects of bladder physiology should be revised.


Asunto(s)
Proteínas Proto-Oncogénicas c-kit/genética , Células Madre/metabolismo , Vejiga Urinaria/metabolismo , Animales , Regulación de la Expresión Génica/genética , Cobayas , Humanos , Células Intersticiales de Cajal/citología , Células Intersticiales de Cajal/metabolismo , Mastocitos/metabolismo , Ratones , Músculo Liso/crecimiento & desarrollo , Músculo Liso/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Ratas , Vejiga Urinaria/citología
12.
Clin Cancer Res ; 23(10): 2528-2541, 2017 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-27780859

RESUMEN

Purpose: Human exportin-1 (XPO1) is the key nuclear-cytoplasmic transport protein that exports different cargo proteins out of the nucleus. Inducing nuclear accumulation of these proteins by inhibiting XPO1 causes cancer cell death. First clinical validation of pharmacological inhibition of XPO1 was obtained with the Selective Inhibitor of Nuclear Export (SINE) compound selinexor (KPT-330) demonstrating activity in phase-II/IIb clinical trials when dosed 1 to 3 times weekly. The second-generation SINE compound KPT-8602 shows improved tolerability and can be dosed daily. Here, we investigate and validate the drug-target interaction of KPT-8602 and explore its activity against acute lymphoblastic leukemia (ALL).Experimental Design: We examined the effect of KPT-8602 on XPO1 function and XPO1-cargo as well as on a panel of leukemia cell lines. Mutant XPO1 leukemia cells were designed to validate KPT-8602's drug-target interaction. In vivo, anti-ALL activity was measured in a mouse ALL model and patient-derived ALL xenograft models.Results: KPT-8602 induced caspase-dependent apoptosis in a panel of leukemic cell lines in vitro Using CRISPR/Cas9 genome editing, we demonstrated the specificity of KPT-8602 for cysteine 528 in the cargo-binding groove of XPO1 and validated the drug target interaction. In vivo, KPT-8602 showed potent anti-leukemia activity in a mouse ALL model as well as in patient-derived T- and B-ALL xenograft models without affecting normal hematopoiesis.Conclusions: KPT-8602 is highly specific for XPO1 inhibition and demonstrates potent anti-leukemic activity supporting clinical application of the second-generation SINE compound for the treatment of ALL. Clin Cancer Res; 23(10); 2528-41. ©2016 AACR.


Asunto(s)
Antineoplásicos/administración & dosificación , Carioferinas/antagonistas & inhibidores , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Tiazoles/administración & dosificación , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Sistemas CRISPR-Cas , Línea Celular Tumoral , Edición Génica , Humanos , Carioferinas/genética , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Receptores Citoplasmáticos y Nucleares/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína Exportina 1
13.
Oncotarget ; 7(42): 68842-68850, 2016 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-27634897

RESUMEN

Exportin-1 (CRM1/XPO1) is a crucial nuclear export protein that transports a wide variety of proteins from the nucleus to the cytoplasm. These cargo proteins include tumor suppressors and growth-regulatory factors and as such XPO1 is considered a potential anti-cancer target. From this perspective, inhibition of the XPO1-mediated nuclear export by selective inhibitor of nuclear export (SINE) compounds has shown broad-spectrum anti-cancer activity. Furthermore, the clinical candidate SINE, selinexor, is currently in multiple phase I/II/IIb trials for treatment of cancer. Resistance against selinexor has not yet been observed in the clinic, but in vitro selection of resistance did not reveal any mutations in the target protein, XPO1. However, introduction of a homozygous mutation at the drug's target site, the cysteine 528 residue inside the XPO1 cargo-binding pocket, by genetic engineering, confers resistance to selinexor. Here we investigated whether this resistance to selinexor is recessive or dominant. For this purpose we have engineered multiple leukemia cell lines containing heterozygous or homozygous C528S substitutions using CRISPR/Cas9-mediated genome editing. Our findings show that heterozygous mutation confers similar resistance against selinexor as homozygous substitution, demonstrating that SINE resistance can be obtained by a single and dominant mutation of the cysteine528 residue in XPO1.


Asunto(s)
Transporte Activo de Núcleo Celular , Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Carioferinas/genética , Receptores Citoplasmáticos y Nucleares/genética , Sistemas CRISPR-Cas , Supervivencia Celular , Cisteína/genética , Edición Génica , Ingeniería Genética , Células HL-60 , Heterocigoto , Homocigoto , Humanos , Células Jurkat , Células K562 , Mutación , Unión Proteica , Proteína Exportina 1
14.
EBioMedicine ; 2(9): 1102-13, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26501108

RESUMEN

Infection with HIV ultimately leads to advanced immunodeficiency resulting in an increased incidence of cancer. For example primary effusion lymphoma (PEL) is an aggressive non-Hodgkin lymphoma with very poor prognosis that typically affects HIV infected individuals in advanced stages of immunodeficiency. Here we report on the dual anti-HIV and anti-PEL effect of targeting a single process common in both diseases. Inhibition of the exportin-1 (XPO1) mediated nuclear transport by clinical stage orally bioavailable small molecule inhibitors (SINE) prevented the nuclear export of the late intron-containing HIV RNA species and consequently potently suppressed viral replication. In contrast, in CRISPR-Cas9 genome edited cells expressing mutant C528S XPO1, viral replication was unaffected upon treatment, clearly demonstrating the anti-XPO1 mechanism of action. At the same time, SINE caused the nuclear accumulation of p53 tumor suppressor protein as well as inhibition of NF-κB activity in PEL cells resulting in cell cycle arrest and effective apoptosis induction. In vivo, oral administration arrested PEL tumor growth in engrafted mice. Our findings provide strong rationale for inhibiting XPO1 as an innovative strategy for the combined anti-retroviral and anti-neoplastic treatment of HIV and PEL and offer perspectives for the treatment of other AIDS-associated cancers and potentially other virus-related malignancies.


Asunto(s)
VIH/efectos de los fármacos , Carioferinas/antagonistas & inhibidores , Linfoma Relacionado con SIDA/tratamiento farmacológico , Terapia Molecular Dirigida , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Acrilatos/química , Acrilatos/farmacología , Acrilatos/uso terapéutico , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Secuencia de Bases , Sistemas CRISPR-Cas/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Femenino , VIH/aislamiento & purificación , Humanos , Carioferinas/metabolismo , Ratones Desnudos , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Unión Proteica/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Reproducibilidad de los Resultados , Triazoles/química , Triazoles/farmacología , Triazoles/uso terapéutico , Proteína p53 Supresora de Tumor/metabolismo , Replicación Viral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/metabolismo , Proteína Exportina 1
15.
Biochem Pharmacol ; 96(3): 190-201, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26070251

RESUMEN

A number of statins, the cholesterol-lowering drugs, inhibit the in vitro replication of hepatitis C virus (HCV). In HCV-infected patients, addition of statins to the earlier standard of care therapy (pegIFN-α and ribavirin) resulted in increased sustained virological response rates. The mechanism by which statins inhibit HCV replication has not yet been elucidated. In an attempt to gain insight in the underlying mechanism, hepatoma cells carrying an HCV replicon were passaged in the presence of increasing concentrations of fluvastatin. Fluvastatin-resistant replicon containing cells could be generated and proved ∼8-fold less susceptible to fluvastatin than wild-type cultures. The growth efficiency of the resistant replicon containing cells was comparable to that of wild-type replicon cells. The fluvastatin-resistant phenotype was not conferred by mutations in the viral genome but is caused by cellular changes. The resistant cell line had a markedly increased HMG-CoA reductase expression upon statin treatment. Furthermore, the expression of the efflux transporter P-gp was increased in fluvastatin-resistant replicon cells (determined by qRT-PCR and flow cytometry). This increased expression resulted also in an increased functional transport activity as measured by the P-gp mediated efflux of calcein AM. In conclusion, we demonstrate that statin resistance in HCV replicon containing hepatoma cells is conferred by changes in the cellular environment.


Asunto(s)
Anticolesterolemiantes/farmacología , Antivirales/farmacología , Ácidos Grasos Monoinsaturados/farmacología , Hepacivirus/efectos de los fármacos , Interacciones Huésped-Patógeno , Indoles/farmacología , Replicación Viral/efectos de los fármacos , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/agonistas , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Carbamatos , Línea Celular Tumoral , Farmacorresistencia Viral , Activación Enzimática , Fluvastatina , Regulación de la Expresión Génica , Hepacivirus/genética , Hepacivirus/crecimiento & desarrollo , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Hepatocitos/virología , Humanos , Hidroximetilglutaril-CoA-Reductasas NADP-Dependientes/genética , Hidroximetilglutaril-CoA-Reductasas NADP-Dependientes/metabolismo , Imidazoles/farmacología , Oligopéptidos/farmacología , Pirrolidinas , Replicón , Transducción de Señal , Valina/análogos & derivados , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/genética , Proteínas Virales/metabolismo
16.
Chem Biol ; 22(1): 107-16, 2015 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-25579209

RESUMEN

Validation of drug-target interaction is essential in drug discovery and development. The ultimate proof for drug-target validation requires the introduction of mutations that confer resistance in cells, an approach that is not straightforward in mammalian cells. Using CRISPR/Cas9 genome editing, we show that a homozygous genomic C528S mutation in the XPO1 gene confers cells with resistance to selinexor (KPT-330). Selinexor is an orally bioavailable inhibitor of exportin-1 (CRM1/XPO1) with potent anticancer activity and is currently under evaluation in human clinical trials. Mutant cells were resistant to the induction of cytotoxicity, apoptosis, cell cycle arrest, and inhibition of XPO1 function, including direct binding of the drug to XPO1. These results validate XPO1 as the prime target of selinexor in cells and identify the selectivity of this drug toward the cysteine 528 residue of XPO1. Our findings demonstrate that CRISPR/Cas9 genome editing enables drug-target validation and drug-target selectivity studies in cancer cells.


Asunto(s)
Antineoplásicos/química , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Hidrazinas/química , Carioferinas/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Triazoles/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Secuencia de Bases , Puntos de Control del Ciclo Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Recombinación Homóloga , Humanos , Hidrazinas/metabolismo , Hidrazinas/farmacología , Células Jurkat , Carioferinas/genética , Carioferinas/metabolismo , Cinética , Mutación , Unión Proteica , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Triazoles/metabolismo , Triazoles/farmacología , Proteína Exportina 1
17.
Antiviral Res ; 112: 91-102, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25453342

RESUMEN

The HIV Rev protein mediates the transport of partially and unspliced HIV mRNA from the nucleus to the cytoplasm. Rev multimerizes on a secondary stem-loop structure present in the viral intron-containing mRNA species and recruits the cellular karyopherin CRM1 to export viral mRNAs from the nucleus to the cytoplasm. Previously we have identified a single-domain intrabody (Nb(190)), derived from a llama heavy-chain antibody, which efficiently inhibits Rev multimerization and suppresses the production of infectious virus. We recently mapped the epitope of this nanobody and demonstrated that Rev residues K20 and Y23 are crucial for interaction while residues V16, H53 and L60 are important to a lesser extent. Here, we generated cell lines stably expressing Nb(190) and assessed the capacity of these cell lines to suppress the replication of different HIV-1 subtypes. These cells stably expressing the single-domain antibody are protected from virus-induced cytopathogenic effect even in the context of high multiplicity of infection. In addition, the replication of different subtypes of group M and one strain of group O is significantly suppressed in these cell lines. Next, we analysed the natural variations of Rev amino acids in sequence samples from HIV-1 infected patients worldwide and assessed the effect of Nb(190) on the most prevalent polymorphisms occurring at the key epitope positions (K20 and Y23) in Rev. We found that Nb(190) was able to suppress the function of these Rev variants except for the K20N mutant, which was present in only 0.7% of HIV-1 sequence populations (n = 4632). Cells stably expressing the single-domain intrabody Nb(190) are protected against virus-induced cytopathogenic effect and display a selective survival advantage upon infection. In addition, Nb(190) suppresses the replication of a wide range of different HIV-1 subtypes. Large-scale sequence analysis reveals that the Nb(190) epitope positions in Rev are well conserved across major HIV-1 subtypes and groups. Altogether, our results indicate that Nb(190) may have broad potential as a gene therapeutic agent against HIV-1.


Asunto(s)
Anticuerpos Anti-VIH/metabolismo , VIH-1/efectos de los fármacos , VIH-1/fisiología , Anticuerpos de Dominio Único/metabolismo , Replicación Viral/efectos de los fármacos , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Animales , Camélidos del Nuevo Mundo , Línea Celular , Efecto Citopatogénico Viral , Expresión Génica , Variación Genética , Anticuerpos Anti-VIH/genética , Humanos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Anticuerpos de Dominio Único/genética , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/genética
18.
J Urol ; 192(5): 1555-63, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24893312

RESUMEN

PURPOSE: There is increasing evidence for an important role of the lamina propria in bladder physiology and interstitial cells seem to have a key role in this area. Interstitial cells in the upper lamina propria have been studied most frequently. We characterized interstitial cells in the deeper lamina propria and hypothesized that the 2 interstitial cell populations have different phenotypes based on their ultrastructural and immunohistochemical properties. MATERIALS AND METHODS: Tissue samples were obtained from macroscopically and microscopically normal areas of radical cystectomy specimens. A panel of immunohistochemical markers was used to characterize lamina propria interstitial cells. Single/double immunohistochemistry/immunofluorescence was performed. At a second phase electron microscopy was used to compare upper and deeper lamina propria interstitial cells. RESULTS: Overall the phenotype of upper lamina propria interstitial cells was vimentin, α-smooth muscle actin, caveolin-1 and 2, PDGFRα, and nonphosphorylated and phosphorylated connexin 43 positive, and CD34 and c-kit negative. The overall phenotype of deeper lamina propria interstitial cells was vimentin, CD34 and nonphosphorylated connexin 43 positive, and α-smooth actin, caveolin-1 and 2, PDGFRα, phosphorylated connexin 43 and c-kit negative. Based on ultrastructural findings upper lamina propria interstitial cells were fibroblasts with myoid features and sparse myofibroblasts while deeper lamina propria interstitial cells were interstitial cell of Cajal-like cells. CONCLUSIONS: To our knowledge this is the first study of 2 main interstitial cell populations in the upper and deeper lamina propria of the human bladder with distinct ultrastructural and immunohistochemical phenotypes. Future research is needed to elucidate whether these morphological findings reflect different roles for upper and deeper lamina propria interstitial cells in bladder physiology.


Asunto(s)
Células Intersticiales de Cajal/metabolismo , Membrana Mucosa/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Vejiga Urinaria/ultraestructura , Anciano , Biomarcadores de Tumor/metabolismo , Caveolina 1/metabolismo , Conexina 43/metabolismo , Femenino , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Humanos , Inmunohistoquímica , Células Intersticiales de Cajal/ultraestructura , Masculino , Microscopía Confocal , Microscopía Electrónica , Membrana Mucosa/ultraestructura , Fenotipo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/metabolismo , Vimentina/metabolismo
19.
J Biol Chem ; 289(18): 12494-506, 2014 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-24634210

RESUMEN

Lens epithelium-derived growth factor (LEDGF/p75) is a transcriptional co-activator involved in targeting human immunodeficiency virus (HIV) integration and the development of MLL fusion-mediated acute leukemia. A previous study revealed that LEDGF/p75 dynamically scans the chromatin, and upon interaction with HIV-1 integrase, their complex is locked on chromatin. At present, it is not known whether LEDGF/p75-mediated chromatin locking is typical for interacting proteins. Here, we employed continuous photobleaching and fluorescence correlation and cross-correlation spectroscopy to investigate in vivo chromatin binding of JPO2, a LEDGF/p75- and c-Myc-interacting protein involved in transcriptional regulation. In the absence of LEDGF/p75, JPO2 performs chromatin scanning inherent to transcription factors. However, whereas the dynamics of JPO2 chromatin binding are decelerated upon interaction with LEDGF/p75, very strong locking of their complex onto chromatin is absent. Similar results were obtained with the domesticated transposase PogZ, another cellular interaction partner of LEDGF/p75. We furthermore show that diffusive JPO2 can oligomerize; that JPO2 and LEDGF/p75 interact directly and specifically in vivo through the specific interaction domain of JPO2 and the C-terminal domain of LEDGF/p75, comprising the integrase-binding domain; and that modulation of JPO2 dynamics requires a functional PWWP domain in LEDGF/p75. Our results suggest that the dynamics of the LEDGF/p75-chromatin interaction depend on the specific partner and that strong chromatin locking is not a property of all LEDGF/p75-binding proteins.


Asunto(s)
Cromatina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas Represoras/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sitios de Unión , Cromatina/genética , Transferencia Resonante de Energía de Fluorescencia , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/genética , Cinética , Microscopía Confocal , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Unión Proteica , Multimerización de Proteína , Proteínas Represoras/química , Proteínas Represoras/genética , Activación Transcripcional , Transposasas/genética , Transposasas/metabolismo
20.
PLoS One ; 8(4): e60259, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23565213

RESUMEN

HIV-1 Rev is the key protein in the nucleocytoplasmic export and expression of the late viral mRNAs. An important aspect for its function is its ability to multimerize on these mRNAs. We have recently identified a llama single-domain antibody (Nb190) as the first inhibitor targeting the Rev multimerization function in cells. This nanobody is a potent intracellular antibody that efficiently inhibits HIV-1 viral production. In order to gain insight into the Nb190-Rev interaction interface, we performed mutational and docking studies to map the interface between the nanobody paratope and the Rev epitope. Alanine mutants of the hyper-variable domains of Nb190 and the Rev multimerization domains were evaluated in different assays measuring Nb190-Rev interaction or viral production. Seven residues within Nb190 and five Rev residues are demonstrated to be crucial for epitope recognition. These experimental data were used to perform docking experiments and map the Nb190-Rev structural interface. This Nb190-Rev interaction model can guide further studies of the Nb190 effect on HIV-1 Rev function and could serve as starting point for the rational development of smaller entities binding to the Nb190 epitope, aimed at interfering with protein-protein interactions of the Rev N-terminal domain.


Asunto(s)
VIH-1/inmunología , Anticuerpos de Dominio Único/inmunología , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/inmunología , Secuencia de Aminoácidos , Fármacos Anti-VIH/química , Fármacos Anti-VIH/inmunología , Fármacos Anti-VIH/farmacología , Afinidad de Anticuerpos/inmunología , Línea Celular , Mapeo Epitopo , Epítopos/inmunología , VIH-1/efectos de los fármacos , VIH-1/genética , Humanos , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Mutación , Unión Proteica/inmunología , Conformación Proteica , Transporte de Proteínas , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/farmacología , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA