Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 11.387
Filtrar
1.
ASAIO J ; 70(6): 546-552, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38829573

RESUMEN

Drug treatments for coronavirus disease 2019 (COVID-19) dramatically improve patient outcomes, and although extracorporeal membrane oxygenation (ECMO) has significant use in these patients, it is unknown whether ECMO affects drug dosing. We used an ex vivo adult ECMO model to measure ECMO circuit effects on concentrations of specific COVID-19 drug treatments. Three identical ECMO circuits used in adult patients were set up. Circuits were primed with fresh human blood (temperature and pH maintained within normal limits). Three polystyrene jars with 75 ml fresh human blood were used as controls. Remdesivir, GS-441524, nafamostat, and tocilizumab were injected in the circuit and control jars at therapeutic concentrations. Samples were taken from circuit and control jars at predefined time points over 6 h and drug concentrations were measured using validated assays. Relative to baseline, mean (± standard deviation [SD]) study drug recoveries in both controls and circuits at 6 h were significantly lower for remdesivir (32.2% [±2.7] and 12.4% [±2.1], p < 0.001), nafamostat (21.4% [±5.0] and 0.0% [±0.0], p = 0.018). Reduced concentrations of COVID-19 drug treatments in ECMO circuits is a clinical concern. Remdesivir and nafamostat may need dose adjustments. Clinical pharmacokinetic studies are suggested to guide optimized COVID-19 drug treatment dosing during ECMO.


Asunto(s)
Adenosina Monofosfato , Alanina , Tratamiento Farmacológico de COVID-19 , Oxigenación por Membrana Extracorpórea , Oxigenación por Membrana Extracorpórea/métodos , Humanos , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/uso terapéutico , Adenosina Monofosfato/farmacocinética , Alanina/análogos & derivados , Alanina/farmacocinética , Alanina/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacocinética , Antivirales/farmacocinética , Antivirales/uso terapéutico , Guanidinas/farmacocinética , Guanidinas/uso terapéutico , Benzamidinas , COVID-19/terapia , SARS-CoV-2 , Adenosina/análogos & derivados
2.
J Vasc Nurs ; 42(2): 138-140, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38823974

RESUMEN

Medical decision-making surrounding high risk surgical procedures requires extensive consideration about the potential risks and benefits to the patient, including implications for concomitant medications and therapies. Managing cardiovascular risk in patients undergoing non-cardiac surgery is essential for safe and effective patient care. In instances where cardiac revascularization is needed prior to surgery, antiplatelet medication is also needed which can complicate future surgical procedures. This case report describes a patient who underwent percutaneous coronary intervention with drug eluting stent placement, who also needed urgent treatment for expanding thoracic abdominal aortic aneurysm (TAAA). Standard practice for endovascular repair of a TAAA includes placement of a lumbar drain to decrease the risk of spinal cord ischemia, however dual antiplatelet therapy is contraindicated. Cangrelor is the only intravenous platelet P2Y12 receptor inhibitor currently available. The use of Cangrelor, a short-acting P2Y12 inhibitor, was successfully utilized as a bridge in the setting of a patient requiring dual antiplatelet therapy (DAPT) and further surgical intervention. This medication may improve outcomes for this subset of patients.


Asunto(s)
Adenosina Monofosfato , Intervención Coronaria Percutánea , Inhibidores de Agregación Plaquetaria , Humanos , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/uso terapéutico , Inhibidores de Agregación Plaquetaria/uso terapéutico , Masculino , Stents Liberadores de Fármacos , Anciano , Aneurisma de la Aorta Abdominal/cirugía , Aneurisma de la Aorta Abdominal/tratamiento farmacológico , Antagonistas del Receptor Purinérgico P2Y/uso terapéutico
3.
Ann Med ; 56(1): 2361843, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38830017

RESUMEN

BACKGROUND: Literature on the safety of remdesivir in hospitalized COVID-19 patients with severe renal impairment is limited. We aimed to investigate the safety and effectiveness of remdesivir in this population. METHODS: We conducted a retrospective cohort study of adult hospitalized COVID-19 patients who received remdesivir between April 2022 and October 2022. Outcomes were compared between estimated glomerular filtration rate (eGFR) <30 mL/min/1.73 m2 and ≥30 mL/min/1.73 m2 groups. The primary safety outcomes were acute kidney injury (AKI) and bradycardia, while the primary effectiveness outcomes included mortality in COVID-19-dedicated wards and hospital mortality. Secondary outcomes included laboratory changes, disease progression, and recovery time. RESULTS: A total of 1,343 patients were recruited, with 307 (22.9%) in the eGFR <30 group and 1,036 (77.1%) in the eGFR ≥30 group. Patients with an eGFR <30 had higher risks of AKI (adjusted hazard ratio [aHR] 2.92, 95% CI 1.93-4.44) and hospital mortality (aHR 1.47, 95% CI 1.06-2.05) but had comparable risks of bradycardia (aHR 1.15, 95% CI 0.85-1.56) and mortality in dedicated wards (aHR 1.43, 95% CI 0.90-2.28) than patients with an eGFR ≥30. Risk of disease progression was higher in the eGFR <30 group (adjusted odds ratio 1.62, 95% CI 1.16-2.26). No difference between the two groups in laboratory changes and recovery time. CONCLUSIONS: Hospitalized COVID-19 patients receiving remdesivir with severe renal impairment had an increased risk of AKI, hospital mortality, and COVID-19 disease progression compared to patients without severe renal impairment.


Asunto(s)
Lesión Renal Aguda , Adenosina Monofosfato , Alanina , Antivirales , Tratamiento Farmacológico de COVID-19 , Tasa de Filtración Glomerular , Mortalidad Hospitalaria , Hospitalización , SARS-CoV-2 , Humanos , Alanina/análogos & derivados , Alanina/uso terapéutico , Alanina/efectos adversos , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/uso terapéutico , Adenosina Monofosfato/efectos adversos , Masculino , Femenino , Estudios Retrospectivos , Persona de Mediana Edad , Anciano , Antivirales/uso terapéutico , Antivirales/efectos adversos , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/epidemiología , Hospitalización/estadística & datos numéricos , COVID-19/complicaciones , COVID-19/mortalidad , Resultado del Tratamiento , Insuficiencia Renal/epidemiología , Bradicardia/inducido químicamente , Bradicardia/epidemiología , Adulto
4.
Sci Rep ; 14(1): 10025, 2024 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-38693137

RESUMEN

The coronavirus-2 has led to a global pandemic of COVID-19 with an outbreak of severe acute respiratory syndrome leading to worldwide quarantine measures and a rise in death rates. The objective of this study is to propose a green, sensitive, and selective densitometric method to simultaneously quantify remdesivir (REM) in the presence of the co-administered drug linezolid (LNZ) and rivaroxaban (RIV) in spiked human plasma. TLC silica gel aluminum plates 60 F254 were used as the stationary phase, and the mobile phase was composed of dichloromethane (DCM): acetone (8.5:1.5, v/v) with densitometric detection at 254 nm. Well-resolved peaks have been observed with retardation factors (Rf) of 0.23, 0.53, and 0.72 for REM, LNZ, and RIV, respectively. A validation study was conducted according to ICH Q2 (R1) Guidelines. The method was rectilinear over the concentration ranges of 0.2-5.5 µg/band, 0.2-4.5 µg/band and 0.1-3.0 µg/band for REM, LNZ and RIV, respectively. The sensitivities of REM, LIN, and RIV were outstanding, with quantitation limits of 128.8, 50.5, and 55.8 ng/band, respectively. The approach has shown outstanding recoveries ranging from 98.3 to 101.2% when applied to pharmaceutical formulations and spiked human plasma. The method's greenness was assessed using Analytical Eco-scale, GAPI, and AGREE metrics.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Antivirales , Tratamiento Farmacológico de COVID-19 , COVID-19 , SARS-CoV-2 , Humanos , Antivirales/sangre , SARS-CoV-2/efectos de los fármacos , COVID-19/sangre , Cromatografía en Capa Delgada/métodos , Análisis Costo-Beneficio , Alanina/sangre , Linezolid/sangre
5.
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
6.
Viruses ; 16(5)2024 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-38793571

RESUMEN

The COVID-19 pandemic has resulted in millions of fatalities worldwide. The case of pediatric cancer patients stands out since, despite being considered a population at risk, few studies have been carried out concerning symptom detection or the description of the mechanisms capable of modifying the course of the COVID-19 disease, such as the interaction and response between the virus and the treatment given to cancer patients. By synthesizing existing studies, this paper aims to expose the treatment challenges for pediatric patients with COVID-19 in an oncology context. Additionally, this updated review includes studies that utilized the antiviral agents Remdesivir and PaxlovidTM in pediatric cancer patients. There is no specific treatment designed exclusively for pediatric cancer patients dealing with COVID-19, and it is advisable to avoid self-medication to prevent potential side effects. Managing COVID-19 in pediatric cancer patients is indeed a substantial challenge. New strategies, such as chemotherapy application rooms, have been implemented for children with cancer who were positive for COVID-19 but asymptomatic since the risk of disease progression is greater than the risk of complications from SARS-CoV-2.


Asunto(s)
Alanina , Antivirales , COVID-19 , Neoplasias , SARS-CoV-2 , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Neoplasias/complicaciones , COVID-19/epidemiología , Niño , Antivirales/uso terapéutico , SARS-CoV-2/efectos de los fármacos , Alanina/análogos & derivados , Alanina/uso terapéutico , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Pandemias
7.
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
9.
Front Immunol ; 15: 1328306, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38590528

RESUMEN

CD39 is the major enzyme controlling the levels of extracellular adenosine triphosphate (ATP) via the stepwise hydrolysis of ATP to adenosine diphosphate (ADP) and adenosine monophosphate (AMP). As extracellular ATP is a strong promoter of inflammation, monoclonal antibodies (mAbs) blocking CD39 are utilized therapeutically in the field of immune-oncology. Though anti-CD39 mAbs are highly specific for their target, they lack deep penetration into the dense tissue of solid tumors, due to their large size. To overcome this limitation, we generated and characterized nanobodies that targeted and blocked human CD39. From cDNA-immunized alpacas we selected 16 clones from seven nanobody families that bind to two distinct epitopes of human CD39. Among these, clone SB24 inhibited the enzymatic activity of CD39. Of note, SB24 blocked ATP degradation by both soluble and cell surface CD39 as a 15kD monomeric nanobody. Dimerization via fusion to an immunoglobulin Fc portion further increased the blocking potency of SB24 on CD39-transfected HEK cells. Finally, we confirmed the CD39 blocking properties of SB24 on human PBMCs. In summary, SB24 provides a new small biological antagonist of human CD39 with potential application in cancer therapy.


Asunto(s)
Anticuerpos de Dominio Único , Humanos , Anticuerpos de Dominio Único/farmacología , Adenosina Trifosfato/metabolismo , Adenosina Monofosfato , Adenosina Difosfato/metabolismo
10.
Sci Rep ; 14(1): 9303, 2024 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-38654033

RESUMEN

Remdesivir (RDV) was the first Food and Drug Administration (FDA)-approved medication for COVID-19, with discordant data on efficacy in reducing mortality risk and disease progression. In the context of a dynamic and rapidly changing pandemic landscape, the utilization of real-world evidence is of utmost importance. The objective of this study is to evaluate the impact of RDV on patients who have been admitted to two university referral hospitals in Italy due to COVID-19. All patients older than 18 years and hospitalized at two different universities (Bari and Palermo) were enrolled in this study. To minimize the effect of potential confounders, we used propensity score matching with one case (Remdesivir) and one control that never experienced this kind of intervention during hospitalization. Mortality was the primary outcome of our investigation, and it was recorded using death certificates and/or medical records. Severe COVID-19 was defined as admission to the intensive care unit or a qSOFAscore ≥ 2 or CURB65scores ≥ 3. After using propensity score matching, 365 patients taking Remdesivir and 365 controls were included. No significant differences emerged between the two groups in terms of mean age and percentage of females, while patients taking Remdesivir were less frequently active smokers (p < 0.0001). Moreover, the patients taking Remdesivir were less frequently vaccinated against COVID-19. All the other clinical, radiological, and pharmacological parameters were balanced between the two groups. The use of Remdesivir in our cohort was associated with a significantly lower risk of mortality during the follow-up period (HR 0.56; 95% CI 0.37-0.86; p = 0.007). Moreover, RDV was associated with a significantly lower incidence of non-invasive ventilation (OR 0.27; 95% CI 0.20-0.36). Furthermore, in the 365 patients taking Remdesivir, we observed two cases of mild renal failure requiring a reduction in the dosage of Remdesivir and two cases in which the physicians decided to interrupt Remdesivir for bradycardia and for QT elongation. Our study suggests that the use of Remdesivir in hospitalized COVID-19 patients is a safe therapy associated with improved clinical outcomes, including halving of mortality and with a reduction of around 75% of the risk of invasive ventilation. In a constantly changing COVID-19 scenario, ongoing research is necessary to tailor treatment decisions based on the latest scientific evidence and optimize patient outcomes.


Asunto(s)
Adenosina Monofosfato , Adenosina Monofosfato/análogos & derivados , Alanina , Alanina/análogos & derivados , Antivirales , Tratamiento Farmacológico de COVID-19 , COVID-19 , Puntaje de Propensión , Humanos , Alanina/uso terapéutico , Adenosina Monofosfato/uso terapéutico , Femenino , Masculino , Italia/epidemiología , Persona de Mediana Edad , Anciano , Antivirales/uso terapéutico , COVID-19/mortalidad , COVID-19/epidemiología , Hospitalización/estadística & datos numéricos , SARS-CoV-2 , Resultado del Tratamiento , Anciano de 80 o más Años , Adulto , Estudios Retrospectivos
12.
Biochem Biophys Res Commun ; 712-713: 149938, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38640739

RESUMEN

Polymerization of nucleotides under prebiotic conditions simulating the early Earth has been extensively studied. Several independent methods have been used to verify that RNA-like polymers can be produced by hot wet-dry cycling of nucleotides. However, it has not been shown that these RNA-like polymers are similar to biological RNA with 3'-5' phosphodiester bonds. In the results described here, RNA-like polymers were generated from 5'-monophosphate nucleosides AMP and UMP. To confirm that the polymers resemble biological RNA, ribonuclease A should catalyze hydrolysis of the 3'-5' phosphodiester bonds between pyrimidine nucleotides to each other or to purine nucleotides, but not purine-purine nucleotide bonds. Here we show AFM images of specific polymers produced by hot wet-dry cycling of AMP, UMP and AMP/UMP (1:1) solutions on mica surfaces, before and after exposure to ribonuclease A. AMP polymers were unaffected by ribonuclease A but UMP polymers disappeared. This indicates that a major fraction of the bonds in the UMP polymers is indeed 3'-5' phosphodiester bonds. Some of the polymers generated from the AMP/UMP mixture also showed clear signs of cleavage. Because ribonuclease A recognizes the ester bonds in the polymers, we show for the first time that these prebiotically produced polymers are in fact similar to biological RNA but are likely to be linked by a mixture of 3'-5' and 2'-5' phosphodiester bonds.


Asunto(s)
ARN , Ribonucleasa Pancreática , ARN/química , ARN/metabolismo , Ribonucleasa Pancreática/química , Ribonucleasa Pancreática/metabolismo , Uridina Monofosfato/química , Uridina Monofosfato/metabolismo , Microscopía de Fuerza Atómica , Calor , Polímeros/química , Adenosina Monofosfato/química , Adenosina Monofosfato/metabolismo , Hidrólisis , Polimerizacion
13.
Nat Commun ; 15(1): 3603, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38684662

RESUMEN

The ability to sense chemical gradients and respond with directional motility and chemical activity is a defining feature of complex living systems. There is a strong interest among scientists to design synthetic systems that emulate these properties. Here, we realize and control such behaviors in a synthetic system by tailoring multivalent interactions of adenosine nucleotides with catalytic microbeads. We first show that multivalent interactions of the bead with gradients of adenosine mono-, di- and trinucleotides (AM/D/TP) control both the phoretic motion and a proton-transfer catalytic reaction, and find that both effects are diminished greatly with increasing valence of phosphates. We exploit this behavior by using enzymatic hydrolysis of ATP to AMP, which downregulates multivalent interactivity in situ. This produces a sudden increase in transport of the catalytic microbeads (a phoretic jump), which is accompanied by increased catalytic activity. Finally, we show how this enzymatic activity can be systematically tuned, leading to simultaneous in situ spatial and temporal control of the location of the microbeads, as well as the products of the reaction that they catalyze. These findings open up new avenues for utilizing multivalent interaction-mediated programming of complex chemo-mechanical behaviors into active systems.


Asunto(s)
Adenosina Trifosfato , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/química , Hidrólisis , Catálisis , Coloides/química , Microesferas , Adenosina Monofosfato/metabolismo , Adenosina Monofosfato/química , Adenosina/metabolismo , Adenosina/química
14.
Nat Commun ; 15(1): 3604, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38684722

RESUMEN

Numerous SARS-CoV-2 variant strains with altered characteristics have emerged since the onset of the COVID-19 pandemic. Remdesivir (RDV), a ribonucleotide analogue inhibitor of viral RNA polymerase, has become a valuable therapeutic agent. However, immunosuppressed hosts may respond inadequately to RDV and develop chronic persistent infections. A patient with respiratory failure caused by interstitial pneumonia, who had undergone transplantation of the left lung, developed COVID-19 caused by Omicron BA.5 strain with persistent chronic viral shedding, showing viral fusogenicity. Genome-wide sequencing analyses revealed the occurrence of several viral mutations after RDV treatment, followed by dynamic changes in the viral populations. The C799F mutation in nsp12 was found to play a pivotal role in conferring RDV resistance, preventing RDV-triphosphate from entering the active site of RNA-dependent RNA polymerase. The occurrence of diverse mutations is a characteristic of SARS-CoV-2, which mutates frequently. Herein, we describe the clinical case of an immunosuppressed host in whom inadequate treatment resulted in highly diverse SARS-CoV-2 mutations that threatened the patient's health due to the development of drug-resistant variants.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Alanina , Alanina/análogos & derivados , COVID-19 , ARN Polimerasa Dependiente de ARN de Coronavirus , Trasplante de Pulmón , Mutación , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , COVID-19/virología , Alanina/uso terapéutico , Masculino , Antivirales/uso terapéutico , Huésped Inmunocomprometido , Adenosina Monofosfato/uso terapéutico , Farmacorresistencia Viral/genética , Persona de Mediana Edad , Tratamiento Farmacológico de COVID-19 , Enfermedades Pulmonares Intersticiales/genética , Enfermedades Pulmonares Intersticiales/virología
15.
Food Chem ; 450: 139343, 2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-38631212

RESUMEN

Ultrasound-assisted freezing (UAF) is a clean technique for meat cryoprotections; however, its effectiveness is still limited compared to conventional cryoprotectants, e.g., sugars, polyols, especially at high dosages. To resolve this problem, a synergistic cryoprotection strategy was developed in this study. Adenosine monophosphate (AMP), an adenosine-type food additive, was introduced into frozen surimi at a considerably reduced content (0.08%), yet substantially enhanced the efficiency of UAF to comparable levels of commercial cryoprotectant (4% sucrose with 4% sorbitol). Specifically, UAF/AMP treatment retarded denaturation of surimi myofibrillar protein (MP) during 60-day frozen storage, as evidenced by its increased solubility, Ca2+-ATPase activity, sulfhydryl content, declined surface hydrophobicity, particle size, and stabilized protein conformation. Gels of UAF/AMP-treated surimi also demonstrated more stabilized microstructures, uniform water distributions, enhanced mechanical properties and water-holding capacities. This study provided a feasible approach to boost the cryoprotective performance of UAF, thus expanding its potential applications in frozen food industry.


Asunto(s)
Adenosina Monofosfato , Crioprotectores , Productos Pesqueros , Congelación , Crioprotectores/química , Crioprotectores/farmacología , Animales , Productos Pesqueros/análisis , Adenosina Monofosfato/química , Conservación de Alimentos/métodos , Conservación de Alimentos/instrumentación , Geles/química , Proteínas de Peces/química , Solubilidad
16.
ACS Chem Biol ; 19(5): 1093-1105, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38646883

RESUMEN

Viral macrodomains that can bind to or hydrolyze protein adenosine diphosphate ribosylation (ADP-ribosylation) have emerged as promising targets for antiviral drug development. Many inhibitor development efforts have been directed against the severe acute respiratory syndrome coronavirus 2 macrodomain 1 (SARS-CoV-2 Mac1). However, potent inhibitors for viral macrodomains are still lacking, with the best inhibitors still in the micromolar range. Based on GS-441524, a remdesivir precursor, and our previous studies, we have designed and synthesized potent binders of SARS-CoV-2 Mac1 and other viral macrodomains including those of Middle East respiratory syndrome coronavirus (MERS-CoV), Venezuelan equine encephalitis virus (VEEV), and Chikungunya virus (CHIKV). We show that the 1'-CN group of GS-441524 promotes binding to all four viral macrodomains tested while capping the 1″-OH of GS-441524-diphosphate-ribose with a simple phenyl ring further contributes to binding. Incorporating these two structural features, the best binders show 20- to 6000-fold increases in binding affinity over ADP-ribose for SARS-CoV-2, MERS-CoV, VEEV, and CHIKV macrodomains. Moreover, building on these potent binders, we have developed two highly sensitive fluorescence polarization tracers that only require nanomolar proteins and can effectively resolve the binding affinities of nanomolar inhibitors. Our findings and probes described here will facilitate future development of more potent viral macrodomain inhibitors.


Asunto(s)
Antivirales , Polarización de Fluorescencia , SARS-CoV-2 , Humanos , Adenosina Difosfato Ribosa/metabolismo , Adenosina Difosfato Ribosa/química , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/química , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/metabolismo , Antivirales/farmacología , Antivirales/química , Antivirales/metabolismo , Virus Chikungunya/efectos de los fármacos , COVID-19/virología , Tratamiento Farmacológico de COVID-19 , Virus de la Encefalitis Equina Venezolana/efectos de los fármacos , Virus de la Encefalitis Equina Venezolana/metabolismo , Coronavirus del Síndrome Respiratorio de Oriente Medio , Unión Proteica , Dominios Proteicos , SARS-CoV-2/efectos de los fármacos
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.
Arch Biochem Biophys ; 756: 109995, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38621448

RESUMEN

T4 polynucleotide kinase (T4 PNK) phosphorylates the 5'-terminus of DNA and RNA substrates. It is widely used in molecular biology. Single nucleotides can serve as substrates if a 3'-phosphate group is present. In this study, the T4 PNK-catalyzed conversion of adenosine 3'-monophosphate (3'-AMP) to adenosine-3',5'-bisphosphate was characterized using isothermal titration calorimetry (ITC). Although ITC is typically used to study ligand binding, in this case the instrument was used to evaluate enzyme kinetics by monitoring the heat production due to reaction enthalpy. The reaction was initiated with a single injection of 3'-AMP substrate into the sample cell containing T4 PNK and ATP at pH 7.6 and 30 °C, and Michaelis-Menten analysis was performed on the reaction rates derived from the plot of differential power versus time. The Michaelis-Menten constant, KM, was 13 µM, and the turnover number, kcat, was 8 s-1. The effect of inhibitors was investigated using pyrophosphate (PPi). PPi caused a dose-dependent decrease in the apparent kcat and increase in the apparent KM under the conditions tested. Additionally, the intrinsic reaction enthalpy and the activation energy of the T4 PNK-catalyzed phosphorylation of 3'-AMP were determined to be -25 kJ/mol and 43 kJ/mol, respectively. ITC is seldom used as a tool to study enzyme kinetics, particularly for technically-challenging enzymes such as kinases. This study demonstrates that quantitative analysis of kinase activity can be amenable to the ITC single injection approach.


Asunto(s)
Calorimetría , Polinucleótido 5'-Hidroxil-Quinasa , Cinética , Calorimetría/métodos , Polinucleótido 5'-Hidroxil-Quinasa/metabolismo , Polinucleótido 5'-Hidroxil-Quinasa/química , Adenosina Monofosfato/química , Adenosina Monofosfato/metabolismo , Termodinámica , Bacteriófago T4/enzimología , Difosfatos/química , Difosfatos/metabolismo , Fosforilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA