Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.228
Filtrar
1.
Open Biol ; 14(6): 230363, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38889796

RESUMEN

We present a novel small molecule antiviral chemotype that was identified by an unconventional cell-free protein synthesis and assembly-based phenotypic screen for modulation of viral capsid assembly. Activity of PAV-431, a representative compound from the series, has been validated against infectious viruses in multiple cell culture models for all six families of viruses causing most respiratory diseases in humans. In animals, this chemotype has been demonstrated efficacious for porcine epidemic diarrhoea virus (a coronavirus) and respiratory syncytial virus (a paramyxovirus). PAV-431 is shown to bind to the protein 14-3-3, a known allosteric modulator. However, it only appears to target the small subset of 14-3-3 which is present in a dynamic multi-protein complex whose components include proteins implicated in viral life cycles and in innate immunity. The composition of this target multi-protein complex appears to be modified upon viral infection and largely restored by PAV-431 treatment. An advanced analog, PAV-104, is shown to be selective for the virally modified target, thereby avoiding host toxicity. Our findings suggest a new paradigm for understanding, and drugging, the host-virus interface, which leads to a new clinical therapeutic strategy for treatment of respiratory viral disease.


Asunto(s)
Antivirales , Antivirales/farmacología , Antivirales/química , Humanos , Animales , Proteínas 14-3-3/metabolismo , Complejos Multiproteicos/metabolismo , Interacciones Huésped-Patógeno/efectos de los fármacos , Línea Celular
2.
Cell Host Microbe ; 32(6): 852-862, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38870901

RESUMEN

Antibiotic resistance, typically associated with genetic changes within a bacterial population, is a frequent contributor to antibiotic treatment failures. Antibiotic persistence and tolerance, which we collectively term recalcitrance, represent transient phenotypic changes in the bacterial population that prolong survival in the presence of typically lethal concentrations of antibiotics. Antibiotic recalcitrance is challenging to detect and investigate-traditionally studied under in vitro conditions, our understanding during infection and its contribution to antibiotic failure is limited. Recently, significant progress has been made in the study of antibiotic-recalcitrant populations in pathogenic species, including Mycobacterium tuberculosis, Staphylococcus aureus, Salmonella enterica, and Yersiniae, in the context of the host environment. Despite the diversity of these pathogens and infection models, shared signals and responses promote recalcitrance, and common features and vulnerabilities of persisters and tolerant bacteria have emerged. These will be discussed here, along with progress toward developing therapeutic interventions to better treat recalcitrant pathogens.


Asunto(s)
Antibacterianos , Bacterias , Humanos , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Bacterias/efectos de los fármacos , Bacterias/genética , Animales , Interacciones Huésped-Patógeno/efectos de los fármacos , Estrés Fisiológico , Farmacorresistencia Bacteriana , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/microbiología , Mycobacterium tuberculosis/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/genética
3.
Int J Biol Macromol ; 270(Pt 2): 132468, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38761900

RESUMEN

The current outbreak of mpox presents a significant threat to the global community. However, the lack of mpox-specific drugs necessitates the identification of additional candidates for clinical trials. In this study, a network medicine framework was used to investigate poxviruses-human interactions to identify potential drugs effective against the mpox virus (MPXV). The results indicated that poxviruses preferentially target hubs on the human interactome, and that these virally-targeted proteins (VTPs) tend to aggregate together within specific modules. Comorbidity analysis revealed that mpox is closely related to immune system diseases. Based on predicted drug-target interactions, 268 drugs were identified using the network proximity approach, among which 23 drugs displaying the least side-effects and significant proximity to MPXV were selected as the final candidates. Lastly, specific drugs were explored based on VTPs, differentially expressed proteins, and intermediate nodes, corresponding to different categories. These findings provide novel insights that can contribute to a deeper understanding of the pathogenesis of MPXV and development of ready-to-use treatment strategies based on drug repurposing.


Asunto(s)
Antivirales , Reposicionamiento de Medicamentos , Reposicionamiento de Medicamentos/métodos , Humanos , Antivirales/farmacología , Mapas de Interacción de Proteínas/efectos de los fármacos , Proteínas Virales , Interacciones Huésped-Patógeno/efectos de los fármacos , Biología Computacional/métodos
4.
Int J Antimicrob Agents ; 63(5): 107138, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38490573

RESUMEN

Intracellular human pathogens are the deadliest infectious diseases and are difficult to treat effectively due to their protection inside the host cell and the development of antimicrobial resistance (AMR). An emerging approach to combat these intracellular pathogens is host-directed therapies (HDT), which harness the innate immunity of host cells. HDT rely on small molecules to promote host protection mechanisms that ultimately lead to pathogen clearance. These therapies are hypothesized to: (1) possess indirect yet broad, cross-species antimicrobial activity, (2) effectively target drug-resistant pathogens, (3) carry a reduced susceptibility to the development of AMR and (4) have synergistic action with conventional antimicrobials. As the field of HDT expands, this systematic review was conducted to collect a compendium of HDT and their characteristics, such as the host mechanisms affected, the pathogen inhibited, the concentrations investigated and the magnitude of pathogen inhibition. The evidential support for the main four HDT hypotheses was assessed and concluded that HDT demonstrate robust cross-species activity, are active against AMR pathogens, clinical isolates and laboratory-adapted pathogens. However, limited information exists to support the notion that HDT are synergistic with canonical antimicrobials and are less predisposed to AMR development.


Asunto(s)
Inmunidad Innata , Humanos , Inmunidad Innata/efectos de los fármacos , Antiinfecciosos/farmacología , Sinergismo Farmacológico , Bacterias/efectos de los fármacos , Antibacterianos/farmacología , Interacciones Huésped-Patógeno/efectos de los fármacos
5.
Plant Dis ; 108(6): 1533-1543, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38105459

RESUMEN

Biopesticide fungicides are naturally derived compounds that offer protection from plant diseases through various modes of action, including antimicrobial activity and upregulation of defense responses in host plants. These plant protectants provide a sustainable and safe alternative to conventional pesticides in integrated disease management programs and are especially salient in the management of high-risk and economically important diseases such as late blight of tomato and potato, for which host resistance options are limited. In this study, a commercially available biopesticide, EF400 comprised of clove (8.2%), rosemary (8.1%), and peppermint oils (6.7%) (Anjon AG, Corcoran, CA), was investigated for its effects on the Phytophthora infestans-tomato pathosystem. Specifically, we evaluated the impact of EF400 on P. infestans growth in culture, disease symptoms in planta, and activation of host defenses through monitoring transcript accumulation of defense-related genes. The application timing and use rate of EF400 were further investigated for managing tomato late blight. EF400 delayed the onset of tomato late blight symptoms, although not as effectively as the copper hydroxide fungicide Champ WG (Nufarm Americas Inc., Alsip, IL). Pathogen mycelial growth and sporangial quantity on late blight-susceptible tomato leaves were significantly reduced with EF400. The biopesticide also had an enhancing or suppressing effect on the transcript accumulation of three defense-related genes: Pin2, PR1a, and PR1b. Our work in exploring a commercially available horticultural oil biopesticide meaningfully contributed to the essential knowledge base for optimizing recommendations for the management of tomato late blight.


Asunto(s)
Phytophthora infestans , Enfermedades de las Plantas , Aceites de Plantas , Solanum lycopersicum , Solanum lycopersicum/microbiología , Phytophthora infestans/efectos de los fármacos , Enfermedades de las Plantas/microbiología , Enfermedades de las Plantas/prevención & control , Aceites de Plantas/farmacología , Fungicidas Industriales/farmacología , Agentes de Control Biológico/farmacología , Mentha piperita/química , Interacciones Huésped-Patógeno/efectos de los fármacos
6.
J Biol Chem ; 299(9): 105119, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37527778

RESUMEN

Serratia marcescens is an opportunistic human pathogen involved in antibiotic-resistant hospital acquired infections. Upon contact with the host epithelial cell and prior to internalization, Serratia induces an early autophagic response that is entirely dependent on the ShlA toxin. Once Serratia invades the eukaryotic cell and multiples inside an intracellular vacuole, ShlA expression also promotes an exocytic event that allows bacterial egress from the host cell without compromising its integrity. Several toxins, including ShlA, were shown to induce ATP efflux from eukaryotic cells. Here, we demonstrate that ShlA triggered a nonlytic release of ATP from Chinese hamster ovary (CHO) cells. Enzymatic removal of accumulated extracellular ATP (eATP) or pharmacological blockage of the eATP-P2Y2 purinergic receptor inhibited the ShlA-promoted autophagic response in CHO cells. Despite the intrinsic ecto-ATPase activity of CHO cells, the effective concentration and kinetic profile of eATP was consistent with the established affinity of the P2Y2 receptor and the known kinetics of autophagy induction. Moreover, eATP removal or P2Y2 receptor inhibition also suppressed the ShlA-induced exocytic expulsion of the bacteria from the host cell. Blocking α5ß1 integrin highly inhibited ShlA-dependent autophagy, a result consistent with α5ß1 transactivation by the P2Y2 receptor. In sum, eATP operates as the key signaling molecule that allows the eukaryotic cell to detect the challenge imposed by the contact with the ShlA toxin. Stimulation of P2Y2-dependent pathways evokes the activation of a defensive response to counteract cell damage and promotes the nonlytic clearance of the pathogen from the infected cell.


Asunto(s)
Autofagia , Interacciones Huésped-Patógeno , Integrina alfa5beta1 , Receptores Purinérgicos P2Y2 , Serratia , Toxinas Biológicas , Animales , Cricetinae , Adenosina Trifosfato/metabolismo , Autofagia/efectos de los fármacos , Células CHO , Cricetulus , Exocitosis/efectos de los fármacos , Interacciones Huésped-Patógeno/efectos de los fármacos , Integrina alfa5beta1/antagonistas & inhibidores , Integrina alfa5beta1/metabolismo , Receptores Purinérgicos P2Y2/metabolismo , Serratia/química , Serratia/efectos de los fármacos , Serratia/fisiología , Toxinas Biológicas/farmacología , Humanos
8.
Proc Natl Acad Sci U S A ; 119(30): e2123065119, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35858407

RESUMEN

SARS-CoV-2, the causative agent of the COVID-19 pandemic, undergoes continuous evolution, highlighting an urgent need for development of novel antiviral therapies. Here we show a quantitative mass spectrometry-based succinylproteomics analysis of SARS-CoV-2 infection in Caco-2 cells, revealing dramatic reshape of succinylation on host and viral proteins. SARS-CoV-2 infection promotes succinylation of several key enzymes in the TCA, leading to inhibition of cellular metabolic pathways. We demonstrated that host protein succinylation is regulated by viral nonstructural protein (NSP14) through interaction with sirtuin 5 (SIRT5); overexpressed SIRT5 can effectively inhibit virus replication. We found succinylation inhibitors possess significant antiviral effects. We also found that SARS-CoV-2 nucleocapsid and membrane proteins underwent succinylation modification, which was conserved in SARS-CoV-2 and its variants. Collectively, our results uncover a regulatory mechanism of host protein posttranslational modification and cellular pathways mediated by SARS-CoV-2, which may become antiviral drug targets against COVID-19.


Asunto(s)
Antivirales , Tratamiento Farmacológico de COVID-19 , COVID-19 , Interacciones Huésped-Patógeno , Terapia Molecular Dirigida , Procesamiento Proteico-Postraduccional , SARS-CoV-2 , Antivirales/farmacología , Antivirales/uso terapéutico , COVID-19/metabolismo , COVID-19/virología , Células CACO-2 , Exorribonucleasas/metabolismo , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , Sirtuinas/metabolismo , Succinatos/metabolismo , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/efectos de los fármacos
9.
Nature ; 603(7899): 25-27, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35233098

Asunto(s)
Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Ensayos Clínicos como Asunto , Reposicionamiento de Medicamentos , Interacciones Huésped-Patógeno/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Adenosina Monofosfato/administración & dosificación , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/uso terapéutico , Administración Oral , Alanina/administración & dosificación , Alanina/análogos & derivados , Alanina/uso terapéutico , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/uso terapéutico , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/economía , Anticuerpos Neutralizantes/uso terapéutico , Antivirales/administración & dosificación , Antivirales/farmacología , COVID-19/economía , COVID-19/inmunología , COVID-19/mortalidad , COVID-19/virología , Vacunas contra la COVID-19 , Citidina/análogos & derivados , Citidina/uso terapéutico , Depsipéptidos/farmacología , Depsipéptidos/uso terapéutico , Dexametasona/administración & dosificación , Dexametasona/uso terapéutico , Combinación de Medicamentos , Sinergismo Farmacológico , Ésteres/farmacología , Ésteres/uso terapéutico , Guanidinas/farmacología , Guanidinas/uso terapéutico , Hospitalización , Humanos , Hidroxilaminas/uso terapéutico , Internacionalidad , Lactamas/uso terapéutico , Leucina/uso terapéutico , Ratones , National Institutes of Health (U.S.)/organización & administración , Nitrilos/uso terapéutico , Factor 1 de Elongación Peptídica/antagonistas & inhibidores , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/uso terapéutico , Prolina/uso terapéutico , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/uso terapéutico , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores
10.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35149555

RESUMEN

SARS-CoV-2 is a highly pathogenic virus that evades antiviral immunity by interfering with host protein synthesis, mRNA stability, and protein trafficking. The SARS-CoV-2 nonstructural protein 1 (Nsp1) uses its C-terminal domain to block the messenger RNA (mRNA) entry channel of the 40S ribosome to inhibit host protein synthesis. However, how SARS-CoV-2 circumvents Nsp1-mediated suppression for viral protein synthesis and if the mechanism can be targeted therapeutically remain unclear. Here, we show that N- and C-terminal domains of Nsp1 coordinate to drive a tuned ratio of viral to host translation, likely to maintain a certain level of host fitness while maximizing replication. We reveal that the stem-loop 1 (SL1) region of the SARS-CoV-2 5' untranslated region (5' UTR) is necessary and sufficient to evade Nsp1-mediated translational suppression. Targeting SL1 with locked nucleic acid antisense oligonucleotides inhibits viral translation and makes SARS-CoV-2 5' UTR vulnerable to Nsp1 suppression, hindering viral replication in vitro at a nanomolar concentration, as well as providing protection against SARS-CoV-2-induced lethality in transgenic mice expressing human ACE2. Thus, SL1 allows Nsp1 to switch infected cells from host to SARS-CoV-2 translation, presenting a therapeutic target against COVID-19 that is conserved among immune-evasive variants. This unique strategy of unleashing a virus' own virulence mechanism against itself could force a critical trade-off between drug resistance and pathogenicity.


Asunto(s)
Regiones no Traducidas 5'/genética , Evasión Inmune/genética , Biosíntesis de Proteínas , SARS-CoV-2/genética , Proteínas no Estructurales Virales/genética , Animales , Secuencia de Bases , Chlorocebus aethiops , Células HEK293 , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Humanos , Evasión Inmune/efectos de los fármacos , Ratones Transgénicos , Modelos Biológicos , Oligonucleótidos Antisentido/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Células Vero , Replicación Viral/efectos de los fármacos
11.
Cell Mol Biol Lett ; 27(1): 10, 2022 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-35109786

RESUMEN

The novel coronavirus disease 2019 (COVID-19) pandemic has spread worldwide, and finding a safe therapeutic strategy and effective vaccine is critical to overcoming severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Therefore, elucidation of pathogenesis mechanisms, especially entry routes of SARS-CoV-2 may help propose antiviral drugs and novel vaccines. Several receptors have been demonstrated for the interaction of spike (S) protein of SARS-CoV-2 with host cells, including angiotensin-converting enzyme (ACE2), ephrin ligands and Eph receptors, neuropilin 1 (NRP-1), P2X7, and CD147. The expression of these entry receptors in the central nervous system (CNS) may make the CNS prone to SARS-CoV-2 invasion, leading to neurodegenerative diseases. The present review provides potential pathological mechanisms of SARS-CoV-2 infection in the CNS, including entry receptors and cytokines involved in neuroinflammatory conditions. Moreover, it explains several neurodegenerative disorders associated with COVID-19. Finally, we suggest inflammasome and JaK inhibitors as potential therapeutic strategies for neurodegenerative diseases.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Sistema Nervioso Central/efectos de los fármacos , Inflamasomas/efectos de los fármacos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Receptores Virales/genética , SARS-CoV-2/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Antivirales/uso terapéutico , Basigina/genética , Basigina/metabolismo , COVID-19/genética , COVID-19/metabolismo , COVID-19/virología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/virología , Efrinas/genética , Efrinas/metabolismo , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Humanos , Factores Inmunológicos/uso terapéutico , Inflamasomas/genética , Inflamasomas/metabolismo , Inhibidores de las Cinasas Janus/uso terapéutico , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/genética , Quinasas Janus/metabolismo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/virología , Neuropilina-1/genética , Neuropilina-1/metabolismo , Receptores Purinérgicos P2X7/genética , Receptores Purinérgicos P2X7/metabolismo , Receptores Virales/antagonistas & inhibidores , Receptores Virales/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad , Transducción de Señal
12.
Life Sci Alliance ; 5(4)2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35110354

RESUMEN

BACKGROUND: There are limited effective prophylactic/early treatments for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Viral entry requires spike protein binding to the angiotensin-converting enzyme-2 receptor and cleavage by transmembrane serine protease 2 (TMPRSS2), a cell surface serine protease. Targeting of TMPRSS2 by either androgen blockade or direct inhibition is in clinical trials in early SARS-CoV-2 infection. METHODS: We used differentiated primary human airway epithelial cells at the air-liquid interface to test the impact of targeting TMPRSS2 on the prevention of SARS-CoV-2 infection. RESULTS: We first modelled the systemic delivery of compounds. Enzalutamide, an oral androgen receptor antagonist, had no impact on SARS-CoV-2 infection. By contrast, camostat mesylate, an orally available serine protease inhibitor, blocked SARS-CoV-2 entry. However, oral camostat is rapidly metabolised in the circulation, with poor airway bioavailability. We therefore modelled local airway administration by applying camostat to the apical surface of differentiated airway cultures. We demonstrated that a brief exposure to topical camostat effectively restricts SARS-CoV-2 infection. CONCLUSION: These experiments demonstrate a potential therapeutic role for topical camostat for pre- or post-exposure prophylaxis of SARS-CoV-2, which can now be evaluated in a clinical trial.


Asunto(s)
Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/virología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , Serina Endopeptidasas/metabolismo , Inhibidores de Serina Proteinasa/administración & dosificación , Administración Tópica , Andrógenos/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Antivirales/farmacología , COVID-19/prevención & control , COVID-19/virología , Células Cultivadas , Células Epiteliales , Ésteres/farmacología , Expresión Génica , Células Caliciformes/inmunología , Células Caliciformes/metabolismo , Guanidinas/farmacología , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Serina Endopeptidasas/genética , Transducción de Señal , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
13.
BMC Plant Biol ; 22(1): 73, 2022 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-35183130

RESUMEN

BACKGROUND: Chitosan has shown potential for the control of Fusarium head blight (FHB) disease caused by Fusarium graminearum. The objective of this study was to compare the effect of chitosan hydrochloride applied pre- or post-fungal inoculation on FHB and to better understand its' mode of action via an untargeted metabolomics study. RESULTS: Chitosan inhibited fungal growth in vitro and, when sprayed on the susceptible wheat cultivar Remus 24 hours pre-inoculation with F. graminearum, it significantly reduced the number of infected spikelets at 7, 14 and 21 days post-inoculation. Chitosan pre-treatment also increased the average grain weight per head, the number of grains per head and the 1000-grain weight compared to the controls sprayed with water. No significant impact of chitosan on grain yield was observed when the plants were sprayed 24 hours post-inoculation with F. graminearum, even if it did result in a reduced number of infected spikelets at every time point. An untargeted metabolomic study using UHPLC-QTOF-MS on wheat spikes revealed that spraying the spikes with both chitosan and F. graminearum activated known FHB resistance pathways (e.g. jasmonic acid). Additionally, more metabolites were up- or down-regulated when both chitosan and F. graminearum spores were sprayed on the spikes (117), as compared with chitosan (51) or F. graminearum on their own (32). This included a terpene, a terpenoid and a liminoid previously associated with FHB resistance. CONCLUSIONS: In this study we showed that chitosan hydrochloride inhibited the spore germination and hyphal development of F. graminearum in vitro, triggered wheat resistance against infection by F. graminearum when used as a pre-inoculant, and highlighted metabolites and pathways commonly and differentially affected by chitosan, the pathogen and both agents. This study provides insights into how chitosan might provide protection or stimulate wheat resistance to infection by F. graminearum. It also unveiled new putatively identified metabolites that had not been listed in previous FHB or chitosan-related metabolomic studies.


Asunto(s)
Quitosano/farmacología , Fusarium/efectos de los fármacos , Enfermedades de las Plantas/microbiología , Triticum/efectos de los fármacos , Triticum/microbiología , Cromatografía Líquida de Alta Presión , Ciclopentanos/metabolismo , Fungicidas Industriales/farmacología , Fusarium/crecimiento & desarrollo , Interacciones Huésped-Patógeno/efectos de los fármacos , Espectrometría de Masas , Metaboloma , Oxilipinas/metabolismo , Triticum/metabolismo
14.
Life Sci ; 295: 120411, 2022 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-35181310

RESUMEN

AIMS: Virus-infected host cells switch their metabolism to a more glycolytic phenotype, required for new virion synthesis and packaging. Therefore, we investigated the effect and mechanistic action of glycolytic inhibitor 2-Deoxy-d-glucose (2-DG) on virus multiplication in host cells following SARS-CoV-2 infection. MAIN METHODS: SARS-CoV-2 induced change in glycolysis was examined in Vero E6 cells. Effect of 2-DG on virus multiplication was evaluated by RT-PCR (N and RdRp genes) analysis, protein expression analysis of Nucleocapsid (N) and Spike (S) proteins and visual indication of cytopathy effect (CPE), The mass spectrometry analysis was performed to examine the 2-DG induced change in glycosylation status of receptor binding domain (RBD) in SARS-CoV-2 spike protein. KEY FINDINGS: We observed SARS-COV-2 infection induced increased glucose influx and glycolysis, resulting in selectively high accumulation of the fluorescent glucose analog, 2-NBDG in Vero E6 cells. 2-DG inhibited glycolysis, reduced virus multiplication and alleviated cells from virus-induced cytopathic effect (CPE) in SARS-CoV-2 infected cells. The progeny virions produced from 2-DG treated cells were found unglycosylated at crucial N-glycosites (N331 and N343) of the receptor-binding domain (RBD) in the spike protein, resulting in production of defective progeny virions with compromised infective potential. SIGNIFICANCE: The mechanistic study revealed that the inhibition of SARS-COV-2 multiplication is attributed to 2-DG induced glycolysis inhibition and possibly un-glycosylation of the spike protein, also. Therefore, based on its previous human trials in different types of Cancer and Herpes patients, it could be a potential molecule to study in COVID-19 patients.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Desoxiglucosa/farmacología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/patogenicidad , Adenosina Trifosfato/metabolismo , Animales , Antivirales/farmacología , COVID-19/metabolismo , COVID-19/virología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Glucosa/metabolismo , Glucólisis/efectos de los fármacos , Glicosilación , Interacciones Huésped-Patógeno/efectos de los fármacos , Manosa/farmacología , SARS-CoV-2/fisiología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Células Vero , Virión/efectos de los fármacos , Virión/patogenicidad , Replicación Viral/efectos de los fármacos
15.
J Biol Chem ; 298(4): 101734, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35181340

RESUMEN

Crop parasites of the Striga genera are a major biological deterrent to food security in Africa and are one of the largest obstacles to poverty alleviation on the continent. Striga seeds germinate by sensing small-molecule hormones, strigolactones (SLs), that emanate from host roots. Although SL receptors (Striga hermonthica HYPOSENSITIVE TO LIGHT [ShHTL]) have been identified, discerning their function has been difficult because these parasites cannot be easily grown under laboratory conditions. Moreover, many Striga species are obligate outcrossers that are not transformable, hence not amenable to genetic analysis. By combining phenotypic screening with ShHTL structural information and hybrid drug discovery methods, we discovered a potent SL perception inhibitor for Striga, dormirazine (DOZ). Structural analysis of this piperazine-based antagonist reveals a novel binding mechanism, distinct from that of known SLs, blocking access of the hormone to its receptor. Furthermore, DOZ reduces the flexibility of protein-protein interaction domains important for receptor signaling to downstream partners. In planta, we show, via temporal additions of DOZ, that SL receptors are required at a specific time during seed conditioning. This conditioning is essential to prime seed germination at the right time; thus, this SL-sensitive stage appears to be critical for adequate receptor signaling. Aside from uncovering a function for ShHTL during seed conditioning, these results suggest that future Ag-Biotech Solutions to Striga infestations will need to carefully time the application of antagonists to exploit receptor availability and outcompete natural SLs, critical elements for successful parasitic plant invasions.


Asunto(s)
Lactonas , Extractos Vegetales , Plantas , Striga , Germinación/efectos de los fármacos , Compuestos Heterocíclicos con 3 Anillos , Interacciones Huésped-Patógeno/efectos de los fármacos , Lactonas/farmacología , Enfermedades de las Plantas/prevención & control , Extractos Vegetales/farmacología , Plantas/parasitología , Striga/efectos de los fármacos , Striga/metabolismo
16.
Sci Adv ; 8(8): eabi6110, 2022 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-35050692

RESUMEN

The spread of SARS-CoV-2 and ongoing COVID-19 pandemic underscores the need for new treatments. Here we report that cannabidiol (CBD) inhibits infection of SARS-CoV-2 in cells and mice. CBD and its metabolite 7-OH-CBD, but not THC or other congeneric cannabinoids tested, potently block SARS-CoV-2 replication in lung epithelial cells. CBD acts after viral entry, inhibiting viral gene expression and reversing many effects of SARS-CoV-2 on host gene transcription. CBD inhibits SARS-CoV-2 replication in part by up-regulating the host IRE1α RNase endoplasmic reticulum (ER) stress response and interferon signaling pathways. In matched groups of human patients from the National COVID Cohort Collaborative, CBD (100 mg/ml oral solution per medical records) had a significant negative association with positive SARS-CoV-2 tests. This study highlights CBD as a potential preventative agent for early-stage SARS-CoV-2 infection and merits future clinical trials. We caution against use of non-medical formulations including edibles, inhalants or topicals as a preventative or treatment therapy at the present time.


Asunto(s)
Antivirales/farmacología , Cannabidiol/farmacología , Interacciones Huésped-Patógeno/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Células A549 , Animales , Antivirales/química , COVID-19/virología , Cannabidiol/química , Cannabidiol/metabolismo , Chlorocebus aethiops , Estrés del Retículo Endoplásmico/efectos de los fármacos , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Células Epiteliales/virología , Femenino , Regulación Viral de la Expresión Génica/efectos de los fármacos , Interacciones Huésped-Patógeno/fisiología , Humanos , Interferones/metabolismo , Ratones , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , SARS-CoV-2/fisiología , Células Vero , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
17.
PLoS Pathog ; 18(1): e1010171, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35025963

RESUMEN

The development of physiological models that reproduce SARS-CoV-2 infection in primary human cells will be instrumental to identify host-pathogen interactions and potential therapeutics. Here, using cell suspensions directly from primary human lung tissues (HLT), we have developed a rapid platform for the identification of viral targets and the expression of viral entry factors, as well as for the screening of viral entry inhibitors and anti-inflammatory compounds. The direct use of HLT cells, without long-term cell culture and in vitro differentiation approaches, preserves main immune and structural cell populations, including the most susceptible cell targets for SARS-CoV-2; alveolar type II (AT-II) cells, while maintaining the expression of proteins involved in viral infection, such as ACE2, TMPRSS2, CD147 and AXL. Further, antiviral testing of 39 drug candidates reveals a highly reproducible method, suitable for different SARS-CoV-2 variants, and provides the identification of new compounds missed by conventional systems, such as VeroE6. Using this method, we also show that interferons do not modulate ACE2 expression, and that stimulation of local inflammatory responses can be modulated by different compounds with antiviral activity. Overall, we present a relevant and rapid method for the study of SARS-CoV-2.


Asunto(s)
Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Pulmón/virología , SARS-CoV-2/fisiología , Internalización del Virus , Adulto , Animales , Antivirales/farmacología , COVID-19/inmunología , COVID-19/patología , Células Cultivadas , Chlorocebus aethiops , Evaluación Preclínica de Medicamentos , Drogas en Investigación/farmacología , Drogas en Investigación/uso terapéutico , Células HEK293 , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Inflamación/patología , Inflamación/terapia , Inflamación/virología , Pulmón/patología , SARS-CoV-2/efectos de los fármacos , Células Vero , Internalización del Virus/efectos de los fármacos
18.
Nat Commun ; 13(1): 121, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013215

RESUMEN

HIV is difficult to eradicate due to the persistence of a long-lived reservoir of latently infected cells. Previous studies have shown that natural killer cells are important to inhibiting HIV infection, but it is unclear whether the administration of natural killer cells can reduce rebound viremia when anti-retroviral therapy is discontinued. Here we show the administration of allogeneic human peripheral blood natural killer cells delays viral rebound following interruption of anti-retroviral therapy in humanized mice infected with HIV-1. Utilizing genetically barcoded virus technology, we show these natural killer cells efficiently reduced viral clones rebounding from latency. Moreover, a kick and kill strategy comprised of the protein kinase C modulator and latency reversing agent SUW133 and allogeneic human peripheral blood natural killer cells during anti-retroviral therapy eliminated the viral reservoir in a subset of mice. Therefore, combinations utilizing latency reversal agents with targeted cellular killing agents may be an effective approach to eradicating the viral reservoir.


Asunto(s)
Fármacos Anti-VIH/farmacología , Linfocitos T CD4-Positivos/inmunología , Infecciones por VIH/terapia , VIH-1/efectos de los fármacos , Células Asesinas Naturales/inmunología , Inhibidores de Proteínas Quinasas/farmacología , Viremia/terapia , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/inmunología , Médula Ósea/virología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/virología , Técnicas de Cocultivo , Femenino , Infecciones por VIH/genética , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/genética , VIH-1/inmunología , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Células Asesinas Naturales/trasplante , Masculino , Ratones , Ratones Transgénicos , Proteína Quinasa C/genética , Proteína Quinasa C/inmunología , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/virología , Carga Viral/efectos de los fármacos , Viremia/genética , Viremia/inmunología , Viremia/virología , Latencia del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
19.
Genome Med ; 14(1): 10, 2022 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-35086559

RESUMEN

BACKGROUND: The COVID-19 pandemic has resulted in 275 million infections and 5.4 million deaths as of December 2021. While effective vaccines are being administered globally, there is still a great need for antiviral therapies as antigenically novel SARS-CoV-2 variants continue to emerge across the globe. Viruses require host factors at every step in their life cycle, representing a rich pool of candidate targets for antiviral drug design. METHODS: To identify host factors that promote SARS-CoV-2 infection with potential for broad-spectrum activity across the coronavirus family, we performed genome-scale CRISPR knockout screens in two cell lines (Vero E6 and HEK293T ectopically expressing ACE2) with SARS-CoV-2 and the common cold-causing human coronavirus OC43. Gene knockdown, CRISPR knockout, and small molecule testing in Vero, HEK293, and human small airway epithelial cells were used to verify our findings. RESULTS: While we identified multiple genes and functional pathways that have been previously reported to promote human coronavirus replication, we also identified a substantial number of novel genes and pathways. The website https://sarscrisprscreens.epi.ufl.edu/ was created to allow visualization and comparison of SARS-CoV2 CRISPR screens in a uniformly analyzed way. Of note, host factors involved in cell cycle regulation were enriched in our screens as were several key components of the programmed mRNA decay pathway. The role of EDC4 and XRN1 in coronavirus replication in human small airway epithelial cells was verified. Finally, we identified novel candidate antiviral compounds targeting a number of factors revealed by our screens. CONCLUSIONS: Overall, our studies substantiate and expand the growing body of literature focused on understanding key human coronavirus-host cell interactions and exploit that knowledge for rational antiviral drug development.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Genoma Viral , Interacciones Huésped-Patógeno/genética , SARS-CoV-2/genética , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , COVID-19/patología , COVID-19/virología , Chlorocebus aethiops , Exorribonucleasas/antagonistas & inhibidores , Exorribonucleasas/genética , Exorribonucleasas/metabolismo , Edición Génica/métodos , Células HEK293 , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas/antagonistas & inhibidores , Proteínas/genética , Proteínas/metabolismo , Interferencia de ARN , ARN Guía de Kinetoplastida/metabolismo , ARN Interferente Pequeño/metabolismo , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , Células Vero , Replicación Viral/genética , Tratamiento Farmacológico de COVID-19
20.
Nat Commun ; 13(1): 558, 2022 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-35091550

RESUMEN

Five New World mammarenaviruses (NWMs) cause life-threatening hemorrhagic fever (HF). Cellular entry by these viruses is mediated by human transferrin receptor 1 (hTfR1). Here, we demonstrate that an antibody (ch128.1/IgG1) which binds the apical domain of hTfR1, potently inhibits infection of attenuated and pathogenic NWMs in vitro. Computational docking of the antibody Fab crystal structure onto the known structure of hTfR1 shows an overlapping receptor-binding region shared by the Fab and the viral envelope glycoprotein GP1 subunit that binds hTfR1, and we demonstrate competitive inhibition of NWM GP1 binding by ch128.1/IgG1 as the principal mechanism of action. Importantly, ch128.1/IgG1 protects hTfR1-expressing transgenic mice against lethal NWM challenge. Additionally, the antibody is well-tolerated and only partially reduces ferritin uptake. Our findings provide the basis for the development of a novel, host receptor-targeted antibody therapeutic broadly applicable to the treatment of HF of NWM etiology.


Asunto(s)
Antígenos CD/metabolismo , Arenaviridae/metabolismo , Fiebre Hemorrágica Americana/metabolismo , Receptores de Transferrina/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Células A549 , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacología , Antígenos CD/inmunología , Arenaviridae/efectos de los fármacos , Arenaviridae/fisiología , Chlorocebus aethiops , Fiebre Hemorrágica Americana/prevención & control , Fiebre Hemorrágica Americana/virología , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Virus Junin/efectos de los fármacos , Virus Junin/fisiología , Ratones Endogámicos C57BL , Ratones Transgénicos , Simulación del Acoplamiento Molecular , Unión Proteica/efectos de los fármacos , Receptores de Transferrina/antagonistas & inhibidores , Receptores de Transferrina/inmunología , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...