Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.970
Filtrar
1.
Int J Mol Sci ; 25(13)2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-39000589

RESUMEN

Mitogen-activated protein kinase kinase 1 (MAPK kinase 1, MEK1) is a key kinase in the mitogen-activated protein kinase (MAPK) signaling pathway. MEK1 mutations have been reported to lead to abnormal activation that is closely related to the malignant growth and spread of various tumors, making it an important target for cancer treatment. Targeting MEK1, four small-molecular drugs have been approved by the FDA, including Trametinib, Cobimetinib, Binimetinib, and Selumetinib. Recently, a study showed that modification with dehydroalanine (Dha) can also lead to abnormal activation of MEK1, which has the potential to promote tumor development. In this study, we used molecular dynamics simulations and metadynamics to explore the mechanism of abnormal activation of MEK1 caused by the Dha modification and predicted the inhibitory effects of four FDA-approved MEK1 inhibitors on the Dha-modified MEK1. The results showed that the mechanism of abnormal activation of MEK1 caused by the Dha modification is due to the movement of the active segment, which opens the active pocket and exposes the catalytic site, leading to sustained abnormal activation of MEK1. Among four FDA-approved inhibitors, only Selumetinib clearly blocks the active site by changing the secondary structure of the active segment from α-helix to disordered loop. Our study will help to explain the mechanism of abnormal activation of MEK1 caused by the Dha modification and provide clues for the development of corresponding inhibitors.


Asunto(s)
Alanina , MAP Quinasa Quinasa 1 , Simulación de Dinámica Molecular , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 1/química , Alanina/análogos & derivados , Alanina/química , Alanina/farmacología , Alanina/metabolismo , Humanos , Dominio Catalítico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/química , Activación Enzimática/efectos de los fármacos , Bencimidazoles/farmacología , Bencimidazoles/química
2.
PLoS One ; 19(7): e0305927, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39024326

RESUMEN

Fibrotic skin diseases, such as keloids, are pathological results of aberrant tissue healing and are characterized by overgrowth of dermal fibroblasts. Remdesivir (RD), an antiviral drug, has been reported to have pharmacological activities in a wide range of fibrotic diseases. However, whether RD function on skin fibrosis remains unclear. Therefore, in our study, we explored the potential effect and mechanisms of RD on skin fibrosis both in vivo and in vitro. As expected, the results demonstrated that RD alleviated BLM-induced skin fibrosis and attenuates the gross weight of keloid tissues in vivo. Further studies suggested that RD suppressed fibroblast activation and autophagy both in vivo and in vitro. In addition, mechanistic research showed that RD attenuated fibroblasts activation by the TGF-ß1/Smad signaling pathway and inhibited fibroblasts autophagy by the PI3K/Akt/mTOR signaling pathway. In summary, our results demonstrate therapeutic potential of RD for skin fibrosis in the future.


Asunto(s)
Adenosina Monofosfato , Alanina , Fibroblastos , Fibrosis , Transducción de Señal , Piel , Factor de Crecimiento Transformador beta1 , Animales , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo , Fibrosis/tratamiento farmacológico , Alanina/análogos & derivados , Alanina/farmacología , Alanina/uso terapéutico , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/metabolismo , Ratones , Piel/efectos de los fármacos , Piel/patología , Piel/metabolismo , Humanos , Autofagia/efectos de los fármacos , Queloide/tratamiento farmacológico , Queloide/metabolismo , Queloide/patología , Antivirales/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Bleomicina , Fosfatidilinositol 3-Quinasas/metabolismo , Masculino , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Smad/metabolismo
3.
Cardiovasc Toxicol ; 24(7): 656-666, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38851664

RESUMEN

Antiviral therapies for treatment of COVID-19 may be associated with significant proarrhythmic potential. In the present study, the potential cardiotoxic side effects of these therapies were evaluated using a Langendorff model of the isolated rabbit heart. 51 hearts of female rabbits were retrogradely perfused, employing a Langendorff-setup. Eight catheters were placed endo- and epicardially to perform an electrophysiology study, thus obtaining cycle length-dependent action potential duration at 90% of repolarization (APD90), QT intervals and dispersion of repolarization. After generating baseline data, the hearts were assigned to four groups: In group 1 (HXC), hearts were treated with 1 µM hydroxychloroquine. Thereafter, 3 µM hydroxychloroquine were infused additionally. Group 2 (HXC + AZI) was perfused with 3 µM hydroxychloroquine followed by 150 µM azithromycin. In group 3 (LOP) the hearts were perfused with 3 µM lopinavir followed by 5 µM and 10 µM lopinavir. Group 4 (REM) was perfused with 1 µM remdesivir followed by 5 µM and 10 µM remdesivir. Hydroxychloroquine- and azithromycin-based therapies have a significant proarrhythmic potential mediated by action potential prolongation and an increase in dispersion. Lopinavir and remdesivir showed overall significantly less pronounced changes in electrophysiology. In accordance with the reported bradycardic events under remdesivir, it significantly reduced the rate of the ventricular escape rhythm.


Asunto(s)
Potenciales de Acción , Antivirales , Preparación de Corazón Aislado , Animales , Conejos , Femenino , Antivirales/farmacología , Antivirales/toxicidad , Potenciales de Acción/efectos de los fármacos , Tratamiento Farmacológico de COVID-19 , Hidroxicloroquina/toxicidad , Hidroxicloroquina/farmacología , Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/fisiopatología , Cardiotoxicidad , Alanina/análogos & derivados , Alanina/farmacología , Frecuencia Cardíaca/efectos de los fármacos , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/toxicidad , Adenosina Monofosfato/farmacología , Corazón/efectos de los fármacos
4.
Int Immunopharmacol ; 137: 112465, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-38878489

RESUMEN

INTRODUCTION: Ulcerative colitis (UC) is a primary culprit of inflammatory bowel disease that entails prompt and effective clinical intervention. Remdesivir (RDV), a broad-spectrum antiviral nucleotide, has been found to exert anti-inflammatory effects in experimental animals. AIM: This study investigates the prospective anti-inflammatory merit of RDV on an experimental model of UC. The role of SIRT6/FoxC1 in regulating colonic cell inflammation and pyroptosis is delineated. METHOD: Rats were challenged with a single intrarectal dose of acetic acid (AA) solution (2 ml; 4 % v/v) to induce colitis. RDV (20 mg/kg, ip) and sulfasalazine (100 mg/kg, po) were administered to rats 14 days before the injection of AA. RESULTS: Administration of RDV ameliorated colonic cell injury and loss as manifested by improvement of severe colon histopathological mutilation and macroscopic damage and disease activity index scores together with restoration of normal colon weight/length ratio. In addition, RDV alleviated colonic inflammatory reactions, thereby curtailing NF-κB activation and the inflammatory cytokines, TNF-α, IL-18, and IL-1ß. Mitigation of colonic oxidative stress and apoptotic reactions were also evident in the setting of RDV treatment. Mechanistically, RDV enhanced the anti-inflammatory cascade, SIRT6/FoxC1, together with curbing the pyroptotic signal, NLRP3/cleaved caspase-1/Gasdermin D-elicited colonic inflammatory cell death. CONCLUSION: This study reveals, for the first time, the anti-inflammatory effect of RDV against experimental UC. Augmenting SIRT6/FoxC1-mediated repression of colonic inflammation and pyroptosis might advocate the colo-protective potential of RDV.


Asunto(s)
Ácido Acético , Adenosina Monofosfato , Alanina , Antiinflamatorios , Colitis Ulcerosa , Colon , Citocinas , Piroptosis , Sirtuinas , Animales , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/patología , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/inmunología , Piroptosis/efectos de los fármacos , Ratas , Masculino , Colon/patología , Colon/efectos de los fármacos , Colon/inmunología , Sirtuinas/metabolismo , Alanina/análogos & derivados , Alanina/uso terapéutico , Alanina/farmacología , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/uso terapéutico , Adenosina Monofosfato/farmacología , Citocinas/metabolismo , Transducción de Señal/efectos de los fármacos , Modelos Animales de Enfermedad , Guanosina Monofosfato , Humanos
5.
Mol Pharmacol ; 106(1): 71-82, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38769019

RESUMEN

Remdesivir (RDV), a broad-spectrum antiviral agent, is often used together with dexamethasone (DEX) for hospitalized COVID-19 patients requiring respiratory support. Potential hepatic adverse drug reaction is a safety concern associated with the use of RDV. We previously reported that DEX cotreatment effectively mitigates RDV-induced hepatotoxicity and reduces elevated serum alanine aminotransferase and aspartate aminotransferase levels in cultured human primary hepatocytes (HPH) and hospitalized COVID-19 patients, respectively. Yet, the precise mechanism behind this protective drug-drug interaction remains largely unknown. Here, we show that through the activation of p38, c-Jun N-terminal kinase (JNK), and extracellular signal-regulated kinases 1 and 2 (ERK1/2) signaling, RDV induces apoptosis (cleavage of caspases 8, 9, and 3), autophagy (increased autophagosome and LC3-II), and mitochondrial damages (decreased membrane potential, respiration, ATP levels, and increased expression of Bax and the released cytosolic cytochrome C) in HPH. Importantly, cotreatment with DEX partially reversed RDV-induced apoptosis, autophagy, and cell death. Mechanistically, DEX deactivates/dephosphorylates p38, JNK, and ERK1/2 signaling by enhancing the expression of dual specificity protein phosphatase 1 (DUSP1), a mitogen-activated protein kinase (MAPK) phosphatase, in a glucocorticoid receptor (GR)-dependent manner. Knockdown of GR in HPH attenuates DEX-mediated DUSP1 induction, MAPK dephosphorylation, as well as protection against RDV-induced hepatotoxicity. Collectively, our findings suggest a molecular mechanism by which DEX modulates the GR-DUSP1-MAPK regulatory axis to alleviate the adverse actions of RDV in the liver. SIGNIFICANCE STATEMENT: The research uncovers the molecular mechanisms by which dexamethasone safeguards against remdesivir-associated liver damage in the context of COVID-19 treatment.


Asunto(s)
Adenosina Monofosfato , Alanina , Antivirales , Apoptosis , Autofagia , Tratamiento Farmacológico de COVID-19 , Enfermedad Hepática Inducida por Sustancias y Drogas , Dexametasona , Fosfatasa 1 de Especificidad Dual , Hepatocitos , Dexametasona/farmacología , Humanos , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Alanina/análogos & derivados , Alanina/farmacología , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Antivirales/farmacología , Antivirales/efectos adversos , Fosfatasa 1 de Especificidad Dual/metabolismo , Fosfatasa 1 de Especificidad Dual/genética , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Células Cultivadas , Sistema de Señalización de MAP Quinasas/efectos de los fármacos
6.
Virol Sin ; 39(3): 459-468, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38782261

RESUMEN

Ebola virus (EBOV) and Marburg virus (MARV), members of the Filoviridae family, are highly pathogenic and can cause hemorrhagic fevers, significantly impacting human society. Bats are considered reservoirs of these viruses because related filoviruses have been discovered in bats. However, due to the requirement for maximum containment laboratories when studying infectious viruses, the characterization of bat filoviruses often relies on pseudoviruses and minigenome systems. In this study, we used RACE technology to sequence the 3'-leader and 5'-trailer of Menglà virus (MLAV) and constructed a minigenome. Similar to MARV, the transcription activities of the MLAV minigenome are independent of VP30. We further assessed the effects of polymorphisms at the 5' end on MLAV minigenome activity and identified certain mutations that decrease minigenome reporter efficiency, probably due to alterations in the RNA secondary structure. The reporter activity upon recombination of the 3'-leaders and 5'-trailers of MLAV, MARV, and EBOV with those of the homologous or heterologous minigenomes was compared and it was found that the polymerase complex and leader and trailer sequences exhibit intrinsic specificities. Additionally, we investigated whether the polymerase complex proteins from EBOV and MARV support MLAV minigenome RNA synthesis and found that the homologous system is more efficient than the heterologous system. Remdesivir efficiently inhibited MLAV as well as EBOV replication. In summary, this study provides new information on bat filoviruses and the minigenome will be a useful tool for high-throughput antiviral drug screening.


Asunto(s)
Ebolavirus , Genoma Viral , Marburgvirus , Animales , Genoma Viral/genética , Ebolavirus/genética , Humanos , Marburgvirus/genética , Mengovirus/genética , Replicación Viral , ARN Viral/genética , Alanina/análogos & derivados , Alanina/farmacología , Quirópteros/virología , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/metabolismo , Filoviridae/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
7.
Microbiol Res ; 285: 127742, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38723390

RESUMEN

In recent years, numerous oomycete mycoviruses have been discovered; however, very few studies have focused on their effects on the host oomycete phenotype. In this study, we investigated the impact of toti-like Pythium ultimum RNA virus 2 (PuRV2) infection on the phytopathogenic soil-borne oomycete Globisporangium ultimum, which serves as a model species for Globisporangium and Pythium, specifically the UOP226 isolate in Japan. We generated a PuRV2-free isogenic line through hyphal tip isolation using high-temperature culture and subsequently compared the phenotypic characteristics and gene expression profiles of UOP226 and the PuRV2-free isogenic line. Our findings revealed that the metalaxyl sensitivity of UOP226 was greater than that of the PuRV2-free isogenic line, whereas the mycelial growth rate and colony morphology remained unchanged in the absence of the fungicide. Furthermore, transcriptome analyses using RNA-seq revealed significant downregulation of ABC-type transporter genes, which are involved in fungicide sensitivity, in UOP226. Our results suggest that PuRV2 infection influences the ecology of G. ultimum in agricultural ecosystems where metalaxyl is applied.


Asunto(s)
Alanina , Virus Fúngicos , Fungicidas Industriales , Enfermedades de las Plantas , Virus ARN , Fungicidas Industriales/farmacología , Virus Fúngicos/genética , Virus Fúngicos/fisiología , Virus Fúngicos/aislamiento & purificación , Virus Fúngicos/efectos de los fármacos , Alanina/análogos & derivados , Alanina/farmacología , Enfermedades de las Plantas/microbiología , Enfermedades de las Plantas/virología , Virus ARN/efectos de los fármacos , Virus ARN/genética , Pythium/efectos de los fármacos , Pythium/crecimiento & desarrollo , Hifa/crecimiento & desarrollo , Hifa/efectos de los fármacos , Perfilación de la Expresión Génica , Micelio/crecimiento & desarrollo , Micelio/efectos de los fármacos , Micelio/virología , Japón , Transcriptoma
8.
Microbiol Res ; 285: 127750, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38761489

RESUMEN

The progress of viral infection involves numerous transcriptional regulatory events. The identification of the newly synthesized transcripts helps us to understand the replication mechanisms and pathogenesis of the virus. Here, we utilized a time-resolved technique called metabolic RNA labeling approach called thiol(SH)-linked alkylation for the metabolic sequencing of RNA (SLAM-seq) to differentially elucidate the levels of steady-state and newly synthesized RNAs of BHK21 cell line in response to human coronavirus OC43 (HCoV-OC43) infection. Our results showed that the Wnt/ß-catenin signaling pathway was significantly enriched with the newly synthesized transcripts of BHK21 cell line in response to HCoV-OC43 infection. Moreover, inhibition of the Wnt pathway promoted viral replication in the early stage of infection, but inhibited it in the later stage of infection. Furthermore, remdesivir inhibits the upregulation of the Wnt/ß-catenin signaling pathway induced by early infection with HCoV-OC43. Collectively, our study showed the diverse roles of Wnt/ß-catenin pathway at different stages of HCoV-OC43 infection, suggesting a potential target for the antiviral treatment. In addition, although infection with HCoV-OC43 induces cytopathic effects in BHK21 cells, inhibiting apoptosis does not affect the intracellular replication of the virus. Monitoring newly synthesized RNA based on such time-resolved approach is a highly promising method for studying the mechanism of viral infections.


Asunto(s)
Adenosina Monofosfato , Alanina , Antivirales , Coronavirus Humano OC43 , Transcriptoma , Replicación Viral , Vía de Señalización Wnt , Coronavirus Humano OC43/genética , Coronavirus Humano OC43/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Línea Celular , Humanos , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/metabolismo , Antivirales/farmacología , Alanina/análogos & derivados , Alanina/farmacología , Alanina/metabolismo , Animales , Infecciones por Coronavirus/virología , Infecciones por Coronavirus/tratamiento farmacológico
9.
J Med Chem ; 67(9): 7470-7486, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38690769

RESUMEN

We assessed factors that determine the tissue-specific bioactivation of ProTide prodrugs by comparing the disposition and activation of remdesivir (RDV), its methylpropyl and isopropyl ester analogues (MeRDV and IsoRDV, respectively), the oral prodrug GS-621763, and the parent nucleotide GS-441524 (Nuc). RDV and MeRDV yielded more active metabolite remdesivir-triphosphate (RDV-TP) than IsoRDV, GS-621763, and Nuc in human lung cell models due to superior cell permeability and higher susceptivity to cathepsin A. Intravenous administration to mice showed that RDV and MeRDV delivered significantly more RDV-TP to the lung than other compounds. Nevertheless, all four ester prodrugs exhibited very low oral bioavailability (<2%), with Nuc being the predominant metabolite in blood. In conclusion, ProTides prodrugs, such as RDV and MeRDV, are more efficient in delivering active metabolites to the lung than Nuc, driven by high cell permeability and susceptivity to cathepsin A. Optimizing ProTides' ester structures is an effective strategy for enhancing prodrug activation in the lung.


Asunto(s)
Adenosina/análogos & derivados , Antivirales , Catepsina A , Pulmón , Profármacos , Profármacos/química , Profármacos/metabolismo , Profármacos/farmacocinética , Profármacos/farmacología , Animales , Ratones , Antivirales/farmacocinética , Antivirales/farmacología , Antivirales/química , Antivirales/metabolismo , Humanos , Catepsina A/metabolismo , Pulmón/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacocinética , Adenosina Monofosfato/metabolismo , Adenosina Monofosfato/química , Adenosina Monofosfato/farmacología , Alanina/análogos & derivados , Alanina/química , Alanina/farmacocinética , Alanina/metabolismo , Alanina/farmacología , Permeabilidad , ProTides
10.
Georgian Med News ; (348): 151-153, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38807409

RESUMEN

Rebamipide contributes to the improvement of blood supply of the GI mucosa, activates its barrier function, activates alkaline secretion of the stomach, increases proliferation and metabolism of epithelial cells of the GI tract, cleanses the mucosa from hydroxyl radicals and suppresses superoxides, produced by polymorphonuclear leukocytes and neutrophils in the presence of Helicobacter pylori, protects the GI mucosa from bacterial invasion and the damaging effects of non-steroidal anti-inflammatory drugs (NSAIDs) on the mucosa. Rebamipide, originally developed as a treatment for gastric ulcers, has attracted the attention of researchers as a potential drug for the treatment of UC due to its ability to stimulate mucus production, reduce oxidative stress, and decrease inflammation. Due to the presence of these properties, it is hypothesized that rebamipide may have a protective effect on the intestinal mucosa during prolonged inflammation, making it a promising candidate for inclusion in therapeutic strategies for ulcerative colitis. The results of this study suggest that rebamipide holds potential therapeutic benefits for the treatment of ulcerative colitis.


Asunto(s)
Alanina , Colitis Ulcerosa , Quinolonas , Quinolonas/uso terapéutico , Quinolonas/farmacología , Alanina/análogos & derivados , Alanina/uso terapéutico , Alanina/farmacología , Animales , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/patología , Ratas , Antiulcerosos/uso terapéutico , Antiulcerosos/farmacología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Mucosa Intestinal/metabolismo , Masculino , Progresión de la Enfermedad , Modelos Animales de Enfermedad , Ratas Wistar
11.
Emerg Microbes Infect ; 13(1): 2353302, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38753462

RESUMEN

Animal models of COVID-19 facilitate the development of vaccines and antivirals against SARS-CoV-2. The efficacy of antivirals or vaccines may differ in different animal models with varied degrees of disease. Here, we introduce a mouse model expressing human angiotensin-converting enzyme 2 (ACE2). In this model, ACE2 with the human cytokeratin 18 promoter was knocked into the Hipp11 locus of C57BL/6J mouse by CRISPR - Cas9 (K18-hACE2 KI). Upon intranasal inoculation with high (3 × 105 PFU) or low (2.5 × 102 PFU) dose of SARS-CoV-2 wildtype (WT), Delta, Omicron BA.1, or Omicron BA.2 variants, all mice showed obvious infection symptoms, including weight loss, high viral loads in the lung, and interstitial pneumonia. 100% lethality was observed in K18-hACE2 KI mice infected by variants with a delay of endpoint for Delta and BA.1, and a significantly attenuated pathogenicity was observed for BA.2. The pneumonia of infected mice was accompanied by the infiltration of neutrophils and pulmonary fibrosis in the lung. Compared with K18-hACE2 Tg mice and HFH4-hACE2 Tg mice, K18-hACE2 KI mice are more susceptible to SARS-CoV-2. In the antivirals test, REGN10933 and Remdesivir had limited antiviral efficacies in K18-hACE2 KI mice upon the challenge of SARS-CoV-2 infections, while Nirmatrelvir, monoclonal antibody 4G4, and mRNA vaccines potently protected the mice from death. Our results suggest that the K18-hACE2 KI mouse model is lethal and stable for SARS-CoV-2 infection, and is practicable and stringent to antiviral development.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Antivirales , COVID-19 , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , SARS-CoV-2 , Animales , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/virología , Ratones , SARS-CoV-2/genética , SARS-CoV-2/inmunología , SARS-CoV-2/efectos de los fármacos , Antivirales/farmacología , Humanos , Pulmón/virología , Pulmón/patología , Tratamiento Farmacológico de COVID-19 , Queratina-18/genética , Carga Viral , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/uso terapéutico , Alanina/análogos & derivados , Alanina/farmacología , Técnicas de Sustitución del Gen , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Femenino
12.
Cells ; 13(10)2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38786105

RESUMEN

HIV infection is an ongoing global health issue, despite increased access to antiretroviral therapy (ART). People living with HIV (PLWH) who are virally suppressed through ART still experience negative health outcomes, including neurocognitive impairment. It is increasingly evident that ART may act independently or in combination with HIV infection to alter the immune state, though this is difficult to disentangle in the clinical population. Thus, these experiments used multiplexed chemokine/cytokine arrays to assess peripheral (plasma) and brain (nucleus accumbens; NAc) expression of immune targets in the presence and absence of ART treatment in the EcoHIV mouse model. The findings identify the effects of EcoHIV infection and of treatment with bictegravir (B), emtricitabine (F), and tenofovir alafenamide (TAF) on the expression of numerous immune targets. In the NAc, this included EcoHIV-induced increases in IL-1α and IL-13 expression and B/F/TAF-induced reductions in KC/CXCL1. In the periphery, EcoHIV suppressed IL-6 and LIF expression, while B/F/TAF reduced IL-12p40 expression. In the absence of ART, IBA-1 expression was negatively correlated with CX3CL1 expression in the NAc of EcoHIV-infected mice. These findings identify distinct effects of ART and EcoHIV infection on peripheral and central immune factors and emphasize the need to consider ART effects on neural and immune outcomes.


Asunto(s)
Infecciones por VIH , Animales , Ratones , Infecciones por VIH/inmunología , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/virología , Emtricitabina/uso terapéutico , Emtricitabina/farmacología , Antirretrovirales/uso terapéutico , Antirretrovirales/farmacología , Modelos Animales de Enfermedad , Masculino , Tenofovir/uso terapéutico , Tenofovir/farmacología , Tenofovir/análogos & derivados , Citocinas/metabolismo , Compuestos Heterocíclicos con 3 Anillos/farmacología , Compuestos Heterocíclicos con 3 Anillos/uso terapéutico , Ratones Endogámicos C57BL , Inmunidad/efectos de los fármacos , Alanina/análogos & derivados , Alanina/uso terapéutico , Alanina/farmacología , Piperazinas/farmacología , Piperazinas/uso terapéutico , Amidas , Piridonas
13.
Org Lett ; 26(19): 4127-4131, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38718303

RESUMEN

Hybrid genome-mining/15N-NMR was used to target compounds containing piperazate (Piz) residues, leading to the discovery of caveamides A (1) and B (2) from Streptomyces sp. strain BE230, isolated from New Rankin Cave (Missouri). Caveamides are highly dynamic molecules containing an unprecedented ß-ketoamide polyketide fragment, two Piz residues, and a new N-methyl-cyclohexenylalanine residue. Caveamide B (2) exhibited nanomolar cytotoxicity against several cancer cell lines and nanomolar antimicrobial activity against MRSA and E. coli.


Asunto(s)
Escherichia coli , Staphylococcus aureus Resistente a Meticilina , Streptomyces , Humanos , Estructura Molecular , Streptomyces/química , Escherichia coli/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Antibacterianos/farmacología , Antibacterianos/química , Antibacterianos/aislamiento & purificación , Pruebas de Sensibilidad Microbiana , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/aislamiento & purificación , Alanina/química , Alanina/farmacología , Alanina/análogos & derivados , Ensayos de Selección de Medicamentos Antitumorales , Péptidos/química , Péptidos/farmacología , Péptidos/aislamiento & purificación , Línea Celular Tumoral , Piridazinas
14.
Bioorg Med Chem Lett ; 107: 129794, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38735344

RESUMEN

Chem-KVL is a tandem repeating peptide, with 14 amino acids that was modified based on a short peptide from a fragment of the human host defense protein chemerin. Chem-KVL increases cationicity and hydrophobicity and shows broad-spectrum antibacterial activity. To determine the molecular determinants of Chem-KVL and whether staple-modified Chem-KVL would improve antibacterial activity and protease stability or decrease cytotoxicity, we combined alanine and stapling scanning, and designed a series of alanine and staple-derived Chem-KVL peptides, termed Chem-A1 to Chem-A14 and SCL-1 to SCL-7. We next examined their antibacterial activity against several gram-positive and gram-negative bacteria, their proteolytic stability, and their cytotoxicity. Ala scanning of Chem-KVL suggested that both the positively charged residues (Lys and Arg) and the hydrophobic residues (Lue and Val) were critical for the antibacterial activities of Chem-KVL peptide. Of note, Chem-A4 was able to remarkably inhibit the growth of gram-positive and gram-negative bacteria when compared to the original peptide. And the antibacterial activities of stapled SCL-4 and SCL-7 were several times higher than those of the linear peptide against gram-positive and gram-negative bacteria. Stapling modification of peptides resulted in increased helicity and protein stability when compared with the linear peptide. These stapled peptides, especially SCL-4 and SCL-7, may serve as the leading compounds for further optimization and antimicrobial therapy.


Asunto(s)
Alanina , Antibacterianos , Bacterias Gramnegativas , Bacterias Grampositivas , Pruebas de Sensibilidad Microbiana , Antibacterianos/farmacología , Antibacterianos/química , Antibacterianos/síntesis química , Bacterias Grampositivas/efectos de los fármacos , Bacterias Gramnegativas/efectos de los fármacos , Alanina/química , Alanina/farmacología , Humanos , Péptidos Antimicrobianos/farmacología , Péptidos Antimicrobianos/química , Péptidos Antimicrobianos/síntesis química , Relación Estructura-Actividad , Mutación , Secuencia de Aminoácidos
15.
Br J Pharmacol ; 181(15): 2636-2654, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38616133

RESUMEN

BACKGROUND AND PURPOSE: There is a need for effective anti-COVID-19 treatments, mainly for individuals at risk of severe disease such as the elderly and the immunosuppressed. Drug repositioning has proved effective in identifying drugs that can find a new application for the control of coronavirus disease, in particular COVID-19. The purpose of the present study was to find synergistic antiviral combinations for COVID-19 based on lethal mutagenesis. EXPERIMENTAL APPROACH: The effect of combinations of remdesivir and ribavirin on the infectivity of SARS-CoV-2 in cell culture has been tested. Viral populations were monitored by ultra-deep sequencing, and the decrease of infectivity as a result of the treatment was measured. KEY RESULTS: Remdesivir and ribavirin exerted a synergistic inhibitory activity against SARS-CoV-2, quantified both by CompuSyn (Chou-Talalay method) and Synergy Finder (ZIP-score model). In serial passage experiments, virus extinction was readily achieved with remdesivir-ribavirin combinations at concentrations well below their cytotoxic 50 value, but not with the drugs used individually. Deep sequencing of treated viral populations showed that remdesivir, ribavirin, and their combinations evoked significant increases of the number of viral mutations and haplotypes, as well as modification of diversity indices that characterize viral quasi-species. CONCLUSION AND IMPLICATIONS: SARS-CoV-2 extinction can be achieved by synergistic combination treatments based on lethal mutagenesis. In addition, the results offer prospects of triple drug treatments for effective SARS-CoV-2 suppression.


Asunto(s)
Adenosina Monofosfato , Alanina , Antivirales , Sinergismo Farmacológico , Ribavirina , SARS-CoV-2 , Alanina/análogos & derivados , Alanina/farmacología , Ribavirina/farmacología , Antivirales/farmacología , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , SARS-CoV-2/efectos de los fármacos , Chlorocebus aethiops , Células Vero , Animales , Humanos , Tratamiento Farmacológico de COVID-19 , COVID-19/virología
16.
Molecules ; 29(8)2024 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-38675600

RESUMEN

The natural pesticide phenazine-1-carboxylic acid (PCA) is known to lack phloem mobility, whereas Metalaxyl is a representative phloem systemic fungicide. In order to endow PCA with phloem mobility and also enhance its antifungal activity, thirty-two phenazine-1-carboxylic acid-N-phenylalanine esters conjugates were designed and synthesized by conjugating PCA with the active structure N-acylalanine methyl ester of Metalaxyl. All target compounds were characterized by 1H NMR, 13C NMR and HRMS. The antifungal evaluation results revealed that several target compounds exhibited moderate to potent antifungal activities against Sclerotinia sclerotiorum, Bipolaris sorokiniana, Phytophthora parasitica, Phytophthora citrophthora. In particular, compound F7 displayed excellent antifungal activity against S. sclerotiorum with an EC50 value of 6.57 µg/mL, which was superior to that of Metalaxyl. Phloem mobility study in castor bean system indicated good phloem mobility for the target compounds F1-F16. Particularly, compound F2 exhibited excellent phloem mobility; the content of compound F2 in the phloem sap of castor bean was 19.12 µmol/L, which was six times higher than Metalaxyl (3.56 µmol/L). The phloem mobility tests under different pH culture solutions verified the phloem translocation of compounds related to the "ion trap" effect. The distribution of the compound F2 in tobacco plants further suggested its ambimobility in the phloem, exhibiting directional accumulation towards the apical growth point and the root. These results provide valuable insights for developing phloem mobility fungicides mediated by exogenous compounds.


Asunto(s)
Alanina , Alanina/análogos & derivados , Fenazinas , Fenazinas/química , Fenazinas/farmacología , Fenazinas/síntesis química , Alanina/química , Alanina/farmacología , Phytophthora/efectos de los fármacos , Antifúngicos/farmacología , Antifúngicos/síntesis química , Antifúngicos/química , Floema/metabolismo , Floema/efectos de los fármacos , Ascomicetos/efectos de los fármacos , Ascomicetos/metabolismo , Fungicidas Industriales/farmacología , Fungicidas Industriales/síntesis química , Fungicidas Industriales/química , Diseño de Fármacos , Ésteres/química , Ésteres/farmacología , Ésteres/síntesis química
17.
Viruses ; 16(4)2024 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-38675889

RESUMEN

Remdesivir (RDV) is a broad-spectrum nucleotide analog prodrug approved for the treatment of COVID-19 in hospitalized and non-hospitalized patients with clinical benefit demonstrated in multiple Phase 3 trials. Here we present SARS-CoV-2 resistance analyses from the Phase 3 SIMPLE clinical studies evaluating RDV in hospitalized participants with severe or moderate COVID-19 disease. The severe and moderate studies enrolled participants with radiologic evidence of pneumonia and a room-air oxygen saturation of ≤94% or >94%, respectively. Virology sample collection was optional in the study protocols. Sequencing and related viral load data were obtained retrospectively from participants at a subset of study sites with local sequencing capabilities (10 of 183 sites) at timepoints with detectable viral load. Among participants with both baseline and post-baseline sequencing data treated with RDV, emergent Nsp12 substitutions were observed in 4 of 19 (21%) participants in the severe study and none of the 2 participants in the moderate study. The following 5 substitutions emerged: T76I, A526V, A554V, E665K, and C697F. The substitutions T76I, A526V, A554V, and C697F had an EC50 fold change of ≤1.5 relative to the wildtype reference using a SARS-CoV-2 subgenomic replicon system, indicating no significant change in the susceptibility to RDV. The phenotyping of E665K could not be determined due to a lack of replication. These data reveal no evidence of relevant resistance emergence and further confirm the established efficacy profile of RDV with a high resistance barrier in COVID-19 patients.


Asunto(s)
Adenosina Monofosfato , Adenosina Monofosfato/análogos & derivados , Alanina , Alanina/análogos & derivados , Antivirales , Tratamiento Farmacológico de COVID-19 , COVID-19 , Farmacorresistencia Viral , SARS-CoV-2 , Carga Viral , Humanos , Alanina/uso terapéutico , Alanina/farmacología , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/uso terapéutico , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/genética , Antivirales/farmacología , Antivirales/uso terapéutico , Carga Viral/efectos de los fármacos , COVID-19/virología , Masculino , Femenino , Estudios Retrospectivos , Persona de Mediana Edad , Índice de Severidad de la Enfermedad
18.
Drug Alcohol Depend ; 259: 111276, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38676968

RESUMEN

BACKGROUND: As nicotine dependence represents a longstanding major public health issue, new nicotine cessation pharmacotherapies are needed. Administration of N-oleoyl glycine (OlGly), an endogenous lipid signaling molecule, prevents nicotine-induced conditioned place preference (CPP) through a peroxisome proliferator-activated receptor-alpha (PPARα) dependent mechanism, and also ameliorated withdrawal signs in nicotine-dependent mice. Pharmacological evidence suggests that the methylated analog of OlGly, N-oleoyl alanine (OlAla), has an increased duration of action and may offer translational benefit. Accordingly, OlAla was assessed in nicotine CPP and dependence assays as well as its pharmacokinetics compared to OlGly. METHODS: ICR female and male mice were tested in nicotine-induced CPP with and without the PPARα antagonist GW6471. OlAla was also assessed in nicotine-dependent mice following removal of nicotine minipumps: somatic withdrawal signs, thermal hyper-nociception and altered affective behavior (i.e., light/dark box). Finally, plasma and brain were collected after administration of OlGly or OlAla and analyzed by high-performance liquid chromatography tandem mass spectrometry. RESULTS: OlAla prevented nicotine-induced CPP, but this effect was not blocked by GW6471. OlAla attenuated somatic and affective nicotine withdrawal signs, but not thermal hyper-nociception in nicotine-dependent mice undergoing withdrawal. OlAla and OlGly showed similar time-courses in plasma and brain. CONCLUSIONS: The observation that both molecules showed similar pharmacokinetics argues against the notion that OlAla offers increased metabolic stability. Moreover, while these structurally similar lipids show efficacy in mouse models of reward and dependence, they reduce nicotine reward through distinct mechanisms.


Asunto(s)
Ratones Endogámicos ICR , Nicotina , Recompensa , Síndrome de Abstinencia a Sustancias , Tabaquismo , Animales , Síndrome de Abstinencia a Sustancias/metabolismo , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Ratones , Masculino , Nicotina/farmacología , Femenino , Tabaquismo/metabolismo , PPAR alfa/metabolismo , Alanina/farmacología , Alanina/análogos & derivados , Ácidos Oléicos/farmacología , Glicina/farmacología , Glicina/análogos & derivados , Aminopiridinas/farmacología , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Oxazoles , Tirosina/análogos & derivados
19.
Science ; 383(6688): eadk6176, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38484056

RESUMEN

Obeldesivir (ODV, GS-5245) is an orally administered prodrug of the parent nucleoside of remdesivir (RDV) and is presently in phase 3 trials for COVID-19 treatment. In this work, we show that ODV and its circulating parent nucleoside metabolite, GS-441524, have similar in vitro antiviral activity against filoviruses, including Marburg virus, Ebola virus, and Sudan virus (SUDV). We also report that once-daily oral ODV treatment of cynomolgus monkeys for 10 days beginning 24 hours after SUDV exposure confers 100% protection against lethal infection. Transcriptomics data show that ODV treatment delayed the onset of inflammation and correlated with antigen presentation and lymphocyte activation. Our results offer promise for the further development of ODV to control outbreaks of filovirus disease more rapidly.


Asunto(s)
Alanina , Antivirales , Ebolavirus , Fiebre Hemorrágica Ebola , Nucleósidos , Profármacos , Animales , Administración Oral , Ebolavirus/efectos de los fármacos , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Fiebre Hemorrágica Ebola/prevención & control , Macaca fascicularis , Nucleósidos/administración & dosificación , Nucleósidos/farmacología , Adenosina Monofosfato/administración & dosificación , Adenosina Monofosfato/farmacología , Alanina/administración & dosificación , Alanina/análogos & derivados , Alanina/farmacología , Profármacos/administración & dosificación , Profármacos/farmacología , Antivirales/administración & dosificación , Antivirales/farmacología
20.
Animal ; 18(2): 101049, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38215677

RESUMEN

Our understanding of metabolic alterations triggered by heat stress is incomplete, which limits the designing of nutritional strategies to mitigate negative productive and health effects. Thus, this study aimed to explore the metabolic responses of heat-stressed dairy cows to dietary supplementation with vitamin D3/Ca and vitamin E/Se. Twelve multiparous Holstein cows were enrolled in a split-plot Latin square design with two distinct vitamin E/Se supplementation levels, either at a low (ESe-, n = 6, 11.1 IU/kg vitamin E and 0.55 mg/kg Se) or a high dose (ESe+, n = 6 223 IU/kg vitamin E and 1.8 mg/kg Se) as the main plot. Treatment subplots, arranged in a replicated 3 × 3 Latin square design, comprised heat challenge (Temperature Humidity Index, THI: 72.0-82.0) supplemented with different levels of vitamin D3/Ca: either low (HS/DCa-, 1 012 IU/kg and 0.73%, respectively) or high (HS/DCa+, 3 764 IU/kg and 0.97%, respectively), and a pair-fed control group in thermoneutrality (THI = 61.0-64.0) receiving the low dose of vitamin D3/Ca (TN). The liquid chromatography-mass spectrometry-based metabolome profile was determined in blood plasma and milk sampled at the beginning (day 0) and end (day 14) of each experimental period. The results were analyzed for the effect of (1) TN vs. HS/ESe-/DCa-, and (2) the vitamin E/Se and vitamin D3/Ca supplementation. No group or group × day effects were detected in the plasma metabolome (false discovery rate, FDR > 0.05), except for triglyceride 52:2 being higher (FDR = 0.03) on day 0 than 14. Taurine, creatinine and butyryl-carnitine showed group × day interactions in the milk metabolome (FDR ≤ 0.05) as creatinine (+22%) and butyryl-carnitine (+190%) were increased (P < 0.01) on day 14, and taurine was decreased (-65%, P < 0.01) on day 14 in the heat stress (HS) cows, compared with day 0. Most compounds were unaffected by vitamin E/Se or vitamin D3/Ca supplementation level or their interaction (FDR > 0.05) in plasma and milk, except for milk alanine which was lower (-69%, FDR = 0.03) in the E/Se+ groups, compared with E/Se-. Our results indicated that HS triggered more prominent changes in the milk than in the plasma metabolome, with consistent results in milk suggesting increased muscle catabolism, as reflected by increased creatinine, alanine and citrulline levels. Supplementing with high levels of vitamin E/Se or vitamin D3/Ca or their combination did not appear to affect the metabolic remodeling triggered by HS.


Asunto(s)
Lactancia , Leche , Femenino , Bovinos , Animales , Leche/metabolismo , Creatinina/análisis , Creatinina/metabolismo , Creatinina/farmacología , Dieta/veterinaria , Calor , Suplementos Dietéticos/análisis , Respuesta al Choque Térmico , Vitamina E , Carnitina/metabolismo , Alanina/análisis , Alanina/metabolismo , Alanina/farmacología , Aminoácidos/metabolismo , Vitamina D/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...