Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Nanobiotechnology ; 22(1): 202, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38658952

RESUMEN

Multi-modal combination therapy is regarded as a promising approach to cancer treatment. Combining chemotherapy and phototherapy is an essential multi-modal combination therapy endeavor. Ivermectin (IVM) is a potent antiparasitic agent identified as having potential antitumor properties. However, the fact that it induces protective autophagy while killing tumor cells poses a challenge to its further application. IR780 iodide (IR780) is a near-infrared (NIR) dye with outstanding photothermal therapy (PTT) and photodynamic therapy (PDT) effects. However, the hydrophobicity, instability, and low tumor uptake of IR780 limit its clinical applications. Here, we have structurally modified IR780 with hydroxychloroquine, an autophagy inhibitor, to synthesize a novel compound H780. H780 and IVM can form H780-IVM nanoparticles (H-I NPs) via self-assembly. Using hyaluronic acid (HA) to modify the H-I NPs, a novel nano-delivery system HA/H780-IVM nanoparticles (HA/H-I NPs) was synthesized for chemotherapy-phototherapy of colorectal cancer (CRC). Under NIR laser irradiation, HA/H-I NPs effectively overcame the limitations of IR780 and IVM and exhibited potent cytotoxicity. In vitro and in vivo experiment results showed that HA/H-I NPs exhibited excellent anti-CRC effects. Therefore, our study provides a novel strategy for CRC treatment that could enhance chemo-phototherapy by modulating autophagy.


Asunto(s)
Autofagia , Neoplasias Colorrectales , Reposicionamiento de Medicamentos , Ivermectina , Nanopartículas , Autofagia/efectos de los fármacos , Animales , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/terapia , Humanos , Ratones , Nanopartículas/química , Ivermectina/farmacología , Ivermectina/química , Línea Celular Tumoral , Indoles/química , Indoles/farmacología , Ratones Endogámicos BALB C , Ratones Desnudos , Fotoquimioterapia/métodos , Antineoplásicos/farmacología , Antineoplásicos/química , Fototerapia/métodos , Ácido Hialurónico/química , Hidroxicloroquina/farmacología , Hidroxicloroquina/química , Terapia Fototérmica/métodos
2.
Int J Mol Sci ; 24(18)2023 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-37762406

RESUMEN

The current study describes the encapsulation of hydroxychloroquine, widely used in traditional medicine due to its diverse pharmacological and medicinal uses, in chitosan nanoparticles (CNPs). This work aims to combine the HCQ drug with CS NPs to generate a novel nanocomposite with improved characteristics and bioavailability. HCQ@CS NPs are roughly shaped like roadways and have a smooth surface with an average size of 159.3 ± 7.1 nm, a PDI of 0.224 ± 0.101, and a zeta potential of +46.6 ± 0.8 mV. To aid in the development of pharmaceutical systems for use in cancer therapy, the binding mechanism and affinity of the interaction between HCQ and HCQ@CS NPs and BSA were examined using stopped-flow and other spectroscopic approaches, supplemented by molecular docking analysis. HCQ and HCQ@CS NPs binding with BSA is driven by a ground-state complex formation that may be accompanied by a non-radiative energy transfer process, and binding constants indicate that HCQ@CS NPs-BSA was more stable than HCQ-BSA. The stopped-flow analysis demonstrated that, in addition to increasing BSA affinity, the nanoformulation HCQ@CS NPS changes the binding process and may open new routes for interaction. Docking experiments verified the development of the HCQ-BSA complex, with HCQ binding to site I on the BSA structure, primarily with the amino acids, Thr 578, Gln 579, Gln 525, Tyr 400, and Asn 404. Furthermore, the nanoformulation HCQ@CS NPS not only increased cytotoxicity against the A549 lung cancer cell line (IC50 = 28.57 ± 1.72 µg/mL) compared to HCQ (102.21 ± 0.67 µg/mL), but also exhibited higher antibacterial activity against both Gram-positive and Gram-negative bacteria when compared to HCQ and chloramphenicol, which is in agreement with the binding constants. The nanoformulation developed in this study may offer a viable therapy option for A549 lung cancer.


Asunto(s)
Quitosano , Neoplasias Pulmonares , Nanopartículas , Humanos , Simulación del Acoplamiento Molecular , Quitosano/química , Hidroxicloroquina/farmacología , Liberación de Fármacos , Antibacterianos , Bacterias Gramnegativas/metabolismo , Bacterias Grampositivas/metabolismo , Nanopartículas/química , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo
3.
Herz ; 48(5): 384-392, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36732468

RESUMEN

BACKGROUND: Hydroxychloroquine (HCQ) is commonly used in the treatment of autoimmune diseases and increases the risk of QT interval prolongation. However, it is unclear how HCQ affects atrial electrophysiology and the risk of atrial fibrillation (AF). METHODS: We quantitatively examined the potential atrial arrhythmogenic effects of HCQ on AF using a computational model of human atrial cardiomyocytes. We measured atrial electrophysiological markers after systematically varying HCQ concentrations. RESULTS: The HCQ concentrations were positively correlated with the action potential duration (APD), resting membrane potential, refractory period, APD alternans threshold, and calcium transient alternans threshold (p < 0.05). By contrast, HCQ concentrations were inversely correlated with the maximum upstroke velocity and calcium transient amplitude (p < 0.05). When the therapeutic concentration (Cmax) of HCQ was applied, HCQ increased APD90 by 1.4% in normal sinus rhythm, 1.8% in wild-type AF, and 2.6% in paired-like homeodomain transcription factor 2 (PITX2)+/- AF, but did not affect the alternans thresholds. The overall in silico results suggest no significant atrial arrhythmogenic effects of HCQ at Cmax, instead implying a potential antiarrhythmic role of low-dose HCQ in AF. However, at an HCQ concentration of fourfold Cmax, a rapid pacing rate of 4 Hz induced prominent APD alternans, particularly in the PITX2+/- AF model. CONCLUSION: Our in silico analysis suggests a potential antiarrhythmic role of low-dose HCQ in AF. Concomitant PITX2 mutations and high-dose HCQ treatments may increase the risk of AF, and this potential genotype/dose-dependent arrhythmogenic effect of HCQ should be investigated further.


Asunto(s)
Fibrilación Atrial , Humanos , Fibrilación Atrial/tratamiento farmacológico , Hidroxicloroquina/farmacología , Calcio/farmacología , Miocitos Cardíacos , Atrios Cardíacos , Antiarrítmicos , Electrofisiología
4.
Metallomics ; 14(10)2022 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-36151967

RESUMEN

Contrasting reports exist in the literature regarding the effect of chloroquine treatment on cellular zinc uptake or secretion. Here, we tested the effect of chloroquine administration in the Drosophila model organism. We show that larvae grown on a diet supplemented with 2.5 mg/ml chloroquine lose up to 50% of their stored zinc and around 10% of their total potassium content. This defect in chloroquine-treated animals correlates with the appearance of abnormal autophagolysosomes in the principal cells of the Malpighian tubules, where zinc storage granules reside. We further show that the reported increase of Fluozin-3 fluorescence following treatment of cells with 300 µM chloroquine for 1 h may not reflect increased zinc accumulation, since a similar treatment in Madin-Darby canine kidney cells results in a 36% decrease in their total zinc content. Thus, chloroquine should not be considered a zinc ionophore. Zinc supplementation plus chloroquine treatment restored zinc content both in vivo and in vitro, without correcting autophagic or other ionic alterations, notably in potassium, associated with the chloroquine treatment. We suggest that chloroquine or hydroxychloroquine administration to patients could reduce intracellular zinc storage pools and be part of the drug's mechanism of action.


Asunto(s)
Drosophila melanogaster , Túbulos de Malpighi , Animales , Cloroquina/farmacología , Perros , Hidroxicloroquina/farmacología , Ionóforos/farmacología , Potasio , Zinc/farmacología
5.
J Trace Elem Med Biol ; 71: 126954, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35190326

RESUMEN

Treatments do not replace vaccinations or restrictions, but are practical, effective, and safe means to help to reduce the fatality associated with COVID-19 infection. While no treatment is available and effective for all the current and future variants of COVID-19, treatments reduce the risk of COVID-19 becoming endemic and reduce mortality and collateral damages. The use of Zinc (Zn) for COVID-19 infection is here reviewed. Zn supplementation may help in prevention as well as during the administration of therapies. Zn supplementation reduces the risks of serious outcomes from Covid19 infection. Evidence also suggests that Zn helps in treatments of COVID-19 infection if taken in conjunction with antiviral drugs. The literature supports the use of Zn, with improvements towards a lower risk ranging from 37% in late treatment, RR 0.63 CI [0.53-0.74], to 78% in sufficiency, RR 0.22 CI [0.05-0.96].


Asunto(s)
COVID-19 , Humanos , Hidroxicloroquina/farmacología , Hidroxicloroquina/uso terapéutico , Cloroquina/farmacología , Cloroquina/uso terapéutico , Ivermectina/farmacología , SARS-CoV-2 , Zinc/uso terapéutico , Zinc/farmacología , Antivirales/farmacología , Antivirales/uso terapéutico , Resultado del Tratamiento , Tratamiento Farmacológico de COVID-19
6.
Int J Mol Sci ; 23(3)2022 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-35163703

RESUMEN

This study aimed to identify potential inhibitors and investigate the mechanism of action on SARS-CoV-2 ACE2 receptors using a molecular modeling study and theoretical determination of biological activity. Hydroxychloroquine was used as a pivot structure and antimalarial analogues of 1,2,4,5 tetraoxanes were used for the construction and evaluation of pharmacophoric models. The pharmacophore-based virtual screening was performed on the Molport® database (~7.9 million compounds) and obtained 313 structures. Additionally, a pharmacokinetic study was developed, obtaining 174 structures with 99% confidence for human intestinal absorption and penetration into the blood-brain barrier (BBB); posteriorly, a study of toxicological properties was realized. Toxicological predictions showed that the selected molecules do not present a risk of hepatotoxicity, carcinogenicity, mutagenicity, and skin irritation. Only 54 structures were selected for molecular docking studies, and five structures showed binding affinity (ΔG) values satisfactory for ACE2 receptors (PDB 6M0J), in which the molecule MolPort-007-913-111 had the best ΔG value of -8.540 Kcal/mol, followed by MolPort-002-693-933 with ΔG = -8.440 Kcal/mol. Theoretical determination of biological activity was realized for 54 structures, and five molecules showed potential protease inhibitors. Additionally, we investigated the Mpro receptor (6M0K) for the five structures via molecular docking, and we confirmed the possible interaction with the target. In parallel, we selected the TopsHits 9 with antiviral potential that evaluated synthetic accessibility for future synthesis studies and in vivo and in vitro tests.


Asunto(s)
Hidroxicloroquina/farmacología , SARS-CoV-2/efectos de los fármacos , Tetraoxanos/farmacología , Antivirales/farmacología , Sitios de Unión , Biología Computacional/métodos , Evaluación Preclínica de Medicamentos/métodos , Humanos , Hidroxicloroquina/análogos & derivados , Simulación del Acoplamiento Molecular/métodos , Simulación de Dinámica Molecular , Inhibidores de Proteasas/farmacología , Unión Proteica/efectos de los fármacos , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/metabolismo , Tratamiento Farmacológico de COVID-19
7.
Eur J Pharmacol ; 915: 174670, 2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-34863995

RESUMEN

Hydroxychloroquine (HCQ) is a derivative of the antimalaria drug chloroquine primarily prescribed for autoimmune diseases. Recent attempts to repurpose HCQ in the treatment of corona virus disease 2019 has raised concerns because of its propensity to prolong the QT-segment on the electrocardiogram, an effect associated with increased pro-arrhythmic risk. Since chirality can affect drug pharmacological properties, we have evaluated the functional effects of the R(-) and S(+) enantiomers of HCQ on six ion channels contributing to the cardiac action potential and on electrophysiological parameters of isolated Purkinje fibers. We found that R(-)HCQ and S(+)HCQ block human Kir2.1 and hERG potassium channels in the 1 µM-100 µM range with a 2-4 fold enantiomeric separation. NaV1.5 sodium currents and CaV1.2 calcium currents, as well as KV4.3 and KV7.1 potassium currents remained unaffected at up to 90 µM. In rabbit Purkinje fibers, R(-)HCQ prominently depolarized the membrane resting potential, inducing autogenic activity at 10 µM and 30 µM, while S(+)HCQ primarily increased the action potential duration, inducing occasional early afterdepolarization at these concentrations. These data suggest that both enantiomers of HCQ can alter cardiac tissue electrophysiology at concentrations above their plasmatic levels at therapeutic doses, and that chirality does not substantially influence their arrhythmogenic potential in vitro.


Asunto(s)
Antimaláricos/química , Antimaláricos/farmacología , Corazón/efectos de los fármacos , Hidroxicloroquina/química , Hidroxicloroquina/farmacología , Canales Iónicos/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Arritmias Cardíacas/inducido químicamente , Electrocardiografía , Técnicas Electrofisiológicas Cardíacas , Canales de Potasio Éter-A-Go-Go , Humanos , Potenciales de la Membrana/efectos de los fármacos , Técnicas de Placa-Clamp , Ramos Subendocárdicos/efectos de los fármacos , Conejos , Estereoisomerismo
8.
Molecules ; 26(24)2021 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-34946543

RESUMEN

COVID-19 is the name of the disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection that occurred in 2019. The virus-host-specific interactions, molecular targets on host cell deaths, and the involved signaling are crucial issues, which become potential targets for treatment. Spike protein, angiotensin-converting enzyme 2 (ACE2), cathepsin L-cysteine peptidase, transmembrane protease serine 2 (TMPRSS2), nonstructural protein 1 (Nsp1), open reading frame 7a (ORF7a), viral main protease (3C-like protease (3CLpro) or Mpro), RNA dependent RNA polymerase (RdRp) (Nsp12), non-structural protein 13 (Nsp13) helicase, and papain-like proteinase (PLpro) are molecules associated with SARS-CoV infection and propagation. SARS-CoV-2 can induce host cell death via five kinds of regulated cell death, i.e., apoptosis, necroptosis, pyroptosis, autophagy, and PANoptosis. The mechanisms of these cell deaths are well established and can be disrupted by synthetic small molecules or natural products. There are a variety of compounds proven to play roles in the cell death inhibition, such as pan-caspase inhibitor (z-VAD-fmk) for apoptosis, necrostatin-1 for necroptosis, MCC950, a potent and specific inhibitor of the NLRP3 inflammasome in pyroptosis, and chloroquine/hydroxychloroquine, which can mitigate the corresponding cell death pathways. However, NF-κB signaling is another critical anti-apoptotic or survival route mediated by SARS-CoV-2. Such signaling promotes viral survival, proliferation, and inflammation by inducing the expression of apoptosis inhibitors such as Bcl-2 and XIAP, as well as cytokines, e.g., TNF. As a result, tiny natural compounds functioning as proteasome inhibitors such as celastrol and curcumin can be used to modify NF-κB signaling, providing a responsible method for treating SARS-CoV-2-infected patients. The natural constituents that aid in inhibiting viral infection, progression, and amplification of coronaviruses are also emphasized, which are in the groups of alkaloids, flavonoids, terpenoids, diarylheptanoids, and anthraquinones. Natural constituents derived from medicinal herbs have anti-inflammatory and antiviral properties, as well as inhibitory effects, on the viral life cycle, including viral entry, replication, assembly, and release of COVID-19 virions. The phytochemicals contain a high potential for COVID-19 treatment. As a result, SARS-CoV-2-infected cell death processes and signaling might be of high efficacy for therapeutic targeting effects and yielding encouraging outcomes.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Muerte Celular/efectos de los fármacos , Descubrimiento de Drogas/métodos , Terapia Molecular Dirigida/métodos , SARS-CoV-2/efectos de los fármacos , Clorometilcetonas de Aminoácidos/farmacología , Antivirales/farmacología , Apoptosis/efectos de los fármacos , Furanos/farmacología , Humanos , Hidroxicloroquina/farmacología , Imidazoles/farmacología , Indenos/farmacología , Indoles/farmacología , Necroptosis/efectos de los fármacos , Fitoquímicos/farmacología , Piroptosis/efectos de los fármacos , SARS-CoV-2/metabolismo , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología , Proteínas Virales/antagonistas & inhibidores
9.
Cell Transplant ; 30: 9636897211057440, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34757864

RESUMEN

The inflammatory response is an obstacle to success in both allogeneic and autologous islet transplantation. In autologous islet transplantation (AIT), however, the recipient is also the donor, permitting pretreatment of donor/recipient for a controlled duration prior to transplantation. We sought to exploit this feature of (AIT) by pretreating donor/recipients with chronic pancreatitis undergoing total pancreatectomy and autologous islet transplantation (TPAIT) to test the hypothesis that peri-transplant treatment with the FDA-approved anti-inflammatory hydroxychloroquine (HCQ) improves graft function. In this randomized placebo-controlled pilot clinical study, patients (n = 6) were treated with oral HCQ for 30 days prior to and 90 days after TPAIT. In vivo islet function was assessed via Mixed Meal Tolerance Testing before HCQ treatment, 6- and 12-months after surgery. In vitro islet bioenergetics were assessed at the time of transplantation via extracellular flux analysis of islet preparation samples from the clinical trial cohort and six additional patients (n = 12). Our study shows that HCQ did not alter clinical endpoints, but HCQ-treated patients showed greater spare respiratory capacity (SRC) compared to samples from control patients (P=0.028). Glycolytic metabolism of islet preparations directly correlated with stimulated C-peptide secretion both before and after TPAIT (P=0.01, R2=0.489 and P=0.03, R2=0.674, respectively), and predicted in vivo islet function better than mitochondrial metabolism of islet preps or islet equivalents infused. Overnight culture of islet preparations altered bioenergetic function, significantly decreasing SRC and maximal respiration (P<0.001). In conclusion, while HCQ did not alter clinical outcomes, it was associated with significantly increased SRC in islet preparations. Bioenergetic analyses of islet preparations suggests that culture should be avoided and that glycolysis may be a more sensitive indicator of in vivo islet function than current metrics, including islet oxygen consumption and islet equivalents infused.


Asunto(s)
Metabolismo Energético/inmunología , Inhibidores Enzimáticos/uso terapéutico , Hidroxicloroquina/uso terapéutico , Trasplante de Islotes Pancreáticos/métodos , Trasplante Autólogo/métodos , Ensayos Clínicos como Asunto , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Hidroxicloroquina/farmacología , Masculino , Proyectos Piloto , Resultado del Tratamiento
10.
BMJ ; 373: n949, 2021 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-33903131

RESUMEN

UPDATES: This is the second version (first update) of the living systematic review, replacing the previous version (available as a data supplement). When citing this paper please consider adding the version number and date of access for clarity. OBJECTIVE: To determine and compare the effects of drug prophylaxis on severe acute respiratory syndrome coronavirus virus 2 (SARS-CoV-2) infection and coronavirus disease 2019 (covid-19). DESIGN: Living systematic review and network meta-analysis (NMA). DATA SOURCES: World Health Organization covid-19 database, a comprehensive multilingual source of global covid-19 literature to 4 March 2022. STUDY SELECTION: Randomised trials in which people at risk of covid-19 were allocated to prophylaxis or no prophylaxis (standard care or placebo). Pairs of reviewers independently screened potentially eligible articles. METHODS: After duplicate data abstraction, we conducted random-effects bayesian network meta-analysis. We assessed risk of bias of the included studies using a modification of the Cochrane risk of bias 2.0 tool and assessed the certainty of the evidence using the grading of recommendations assessment, development and evaluation (GRADE) approach. RESULTS: The second iteration of this living NMA includes 32 randomised trials which enrolled 25 147 participants and addressed 21 different prophylactic drugs; adding 21 trials (66%), 18 162 participants (75%) and 16 (76%) prophylactic drugs. Of the 16 prophylactic drugs analysed, none provided convincing evidence of a reduction in the risk of laboratory confirmed SARS-CoV-2 infection. For admission to hospital and mortality outcomes, no prophylactic drug proved different than standard care or placebo. Hydroxychloroquine and vitamin C combined with zinc probably increase the risk of adverse effects leading to drug discontinuation­risk difference for hydroxychloroquine (RD) 6 more per 1000 (95% credible interval (CrI) 2 more to 10 more); for vitamin C combined with zinc, RD 69 more per 1000 (47 more to 90 more), moderate certainty evidence. CONCLUSIONS: Much of the evidence remains very low certainty and we therefore anticipate future studies evaluating drugs for prophylaxis may change the results for SARS-CoV-2 infection, admission to hospital and mortality outcomes. Both hydroxychloroquine and vitamin C combined with zinc probably increase adverse effects. SYSTEMATIC REVIEW REGISTRATION: This review was not registered. The protocol established a priori is included as a supplement. FUNDING: This study was supported by the Canadian Institutes of Health Research (grant CIHR-IRSC:0579001321).


Asunto(s)
COVID-19 , Carragenina/farmacología , Salud Global/estadística & datos numéricos , Hidroxicloroquina/farmacología , Ivermectina/farmacología , Antiinfecciosos/farmacología , COVID-19/prevención & control , Quimioprevención/métodos , Quimioprevención/estadística & datos numéricos , Humanos , SARS-CoV-2 , Resultado del Tratamiento , Incertidumbre
11.
J Nat Prod ; 84(4): 1261-1270, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33844528

RESUMEN

The coronaviruses disease 2019 (COVID-19) caused by a novel coronavirus (SARS-CoV-2) has become a major health problem, affecting more than 50 million people with over one million deaths globally. Effective antivirals are still lacking. Here, we optimized a high-content imaging platform and the plaque assay for viral output study using the legitimate model of human lung epithelial cells, Calu-3, to determine the anti-SARS-CoV-2 activity of Andrographis paniculata extract and its major component, andrographolide. SARS-CoV-2 at 25TCID50 was able to reach the maximal infectivity of 95% in Calu-3 cells. Postinfection treatment of A. paniculata and andrographolide in SARS-CoV-2-infected Calu-3 cells significantly inhibited the production of infectious virions with an IC50 of 0.036 µg/mL and 0.034 µM, respectively, as determined by the plaque assay. The cytotoxicity profile developed over the cell line representatives of major organs, including liver (HepG2 and imHC), kidney (HK-2), intestine (Caco-2), lung (Calu-3), and brain (SH-SY5Y), showed a CC50 of >100 µg/mL for A. paniculata extract and 13.2-81.5 µM for andrographolide, respectively, corresponding to a selectivity index of over 380. In conclusion, this study provided experimental evidence in favor of A. paniculata and andrographolide for further development as a monotherapy or in combination with other effective drugs against SARS-CoV-2 infection.


Asunto(s)
Andrographis , Diterpenos/farmacología , Extractos Vegetales/farmacología , SARS-CoV-2/efectos de los fármacos , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Epiteliales/virología , Humanos , Hidroxicloroquina/farmacología , Pulmón/virología
12.
Virology ; 555: 10-18, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33421743

RESUMEN

Novel coronavirus (SARS-CoV-2), turned out to be a global pandemic with unstoppable morbidity and mortality rate. However, till date there is no effective treatment found against SARS-CoV-2. We report on the major in-depth molecular and docking analysis by using antiretroviral (Lopinavir and ritonavir), antimalarial (Hydroxychloroquine), antibiotics (Azithromycin), and dietary supplements (Vitamin C and E) to provide new insight into drug repurposing molecular events involved in SARS-CoV-2. We constructed three drug-target-pathways-disease networks to predict the targets and drugs interactions as well as important pathways involved in SARS-CoV-2. The results suggested that by using the combination of Lopinavir, Ritonavir along with Hydroxychloroquine and Vitamin C may turned out to be the effective line of treatment for SARS-CoV-2 as it shows the involvement of PARP-1, MAPK-8, EGFR, PRKCB, PTGS-2, and BCL-2. Gene ontology biological process analysis further confirmed multiple viral infection-related processes (P < 0.001), including viral life cycle, modulation by virus, C-C chemokine receptor activity, and platelet activation. KEGG pathway analysis involves multiple pathways (P < 0.05), including FoxO, GnRH, ErbB, Neurotrophin, Toll-like receptor, IL-17, TNF, Insulin, HIF-1, JAK-STAT, Estrogen, NF-kappa, Chemokine, VEGF, and Thyroid hormone signaling pathway in SARS-CoV-2. Docking study was carried out to predict the molecular mechanism Thus, the potential drug combinations could reduce viral infectivity, viral replication, and abnormal host inflammatory responses and may be useful for multi-target drugs against SARS-CoV-2.


Asunto(s)
Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Reposicionamiento de Medicamentos , SARS-CoV-2/efectos de los fármacos , Antivirales/metabolismo , Antivirales/uso terapéutico , Ácido Ascórbico/metabolismo , Ácido Ascórbico/farmacología , Ácido Ascórbico/uso terapéutico , COVID-19/virología , Desarrollo de Medicamentos , Quimioterapia Combinada , Humanos , Hidroxicloroquina/metabolismo , Hidroxicloroquina/farmacología , Hidroxicloroquina/uso terapéutico , Lopinavir/metabolismo , Lopinavir/farmacología , Lopinavir/uso terapéutico , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Ritonavir/metabolismo , Ritonavir/farmacología , Ritonavir/uso terapéutico , SARS-CoV-2/genética , SARS-CoV-2/fisiología , Transducción de Señal , Replicación Viral/efectos de los fármacos
13.
Biomed Pharmacother ; 133: 110891, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33227700

RESUMEN

Severe acute respiratory syndrome (SARS) develops within 3-14 days when CoV2 invades epithelial, myeloid cells in the nasopharynx and pneumocytes in the respiratory tract through angiotensin converting enzyme (ACE2). Infection swiftly disseminates to gastrointestinal, cardiovascular, renal organs as well as immune system to deregulate their normal functioning through unique and distinct mechanisms. The health system and economy has been intensely thwarted by the rapid spread and exorbitant mortality caused by COVID-19 disease across the globe. The acute progression of the disease and high infection rate pose an enormous challenge for its therapeutic management and critical care. The viral structure, genome and proteome have been deciphered which yielded cues for targeting already available therapeutic entities. More than 200 compounds have been screened and till date approximately 69 therapeutic agents are undergoing clinical trials across the world. Among these, remedesivir (RMD), chloroquine (CQ), hydroxychloroquine (HCQ), noscapine (NOS) and heparin have demonstrated fairly promising results in preclinical and clinical studies. Recently, RMD has been approved by USFDA for the management of COVID 19. However, intense research is going on to screen and ace the 'magic bullets' for the management of SARS-CoV2 infection worldwide. The current review illustrates the plausible therapeutic targets in SARS-CoV2 important for inhibition of virus cycle. In addition, the role of RMD, CQ, HCQ, NOS and heparin in combating infection has been addressed. The importance of vitamin C and D supplements as adjunct therapies in the prevention of SARS-CoV2 virus infection have also been summarized.


Asunto(s)
Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Reposicionamiento de Medicamentos , COVID-19/transmisión , Cloroquina/farmacología , Humanos , Hidroxicloroquina/farmacología , Inmunoterapia , SARS-CoV-2/efectos de los fármacos
14.
Phytomedicine ; 79: 153333, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32920291

RESUMEN

BACKGROUND: The novel coronavirus disease (2019-nCoV) has been affecting global health since the end of 2019 and there is no sign that the epidemic is abating . The major issue for controlling the infectious is lacking efficient prevention and therapeutic approaches. Chloroquine (CQ) and Hydroxychloroquine (HCQ) have been reported to treat the disease, but the underlying mechanism remains controversial. PURPOSE: The objective of this study is to investigate whether CQ and HCQ could be ACE2 blockers and used to inhibit 2019-nCoV virus infection. METHODS: In our study, we used CCK-8 staining, flow cytometry and immunofluorescent staining to evaluate the toxicity and autophagy of CQ and HCQ, respectively, on ACE2 high-expressing HEK293T cells (ACE2h cells). We further analyzed the binding character of CQ and HCQ to ACE2 by molecular docking and surface plasmon resonance (SPR) assays, 2019-nCoV spike pseudotyped virus was also used to observe the viropexis effect of CQ and HCQ in ACE2h cells. RESULTS: Results showed that HCQ is slightly more toxic to ACE2h cells than CQ. Both CQ and HCQ could bind to ACE2 with KD = (7.31 ± 0.62)e-7 M and (4.82 ± 0.87)e-7 M, respectively. They exhibit equivalent suppression effect for the entrance of 2019-nCoV spike pseudotyped virus into ACE2h cells. CONCLUSIONS: CQ and HCQ both inhibit the entrance 2019-nCoV into cells by blocking the binding of the virus with ACE2. Our findings provide novel insights into the molecular mechanism of CQ and HCQ treatment effect on virus infection.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Betacoronavirus/efectos de los fármacos , Cloroquina/farmacología , Hidroxicloroquina/farmacología , Peptidil-Dipeptidasa A/efectos de los fármacos , Enzima Convertidora de Angiotensina 2 , Autofagia/efectos de los fármacos , Betacoronavirus/fisiología , COVID-19 , Infecciones por Coronavirus/tratamiento farmacológico , Células HEK293 , Humanos , Simulación del Acoplamiento Molecular , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral , SARS-CoV-2 , Tratamiento Farmacológico de COVID-19
15.
Phytother Res ; 34(12): 3400-3410, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32779305

RESUMEN

The world is in an immediate need of treatment for coronavirus disease (COVID-19). Chronic exposure of hydroxychloroquine in the treatment of COVID-19 may have multiple adverse effects on human physiology, such as cardiac arrhythmias. Natural compounds need to be evaluated as treatment and preventive agents in coronavirus infection. A total of 30 compounds of Solanum tuberosum and Brassica juncea residue smoke water were selected for the virtual screening against SARS-CoV-1, SARS-CoV-2 and cellular proteins involved in the mechanism of infection. Docking analysis identified lead molecules with favorable binding energy, number of poses and hydrogen bond interactions, which indicates the effective modulation of ACE2 and TMPRSS2 receptors. Results indicated (a) curcumenol, (b) N-desmethylselegiline, (c) phentermine and (d) sphingolipid derivatives as a selective and potent candidates in comparison to hydroxychloroquine for COVID-19 treatment. Our in silico findings, therefore, warrant further in vitro validations of the selected compounds for the discovery of novel preventive and therapeutic drug against SARS-CoV-2 infection.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Antivirales/farmacología , Planta de la Mostaza , Fitoquímicos/farmacología , Serina Endopeptidasas/metabolismo , Humo , Solanum tuberosum , Simulación por Computador , Hidroxicloroquina/farmacología , Simulación del Acoplamiento Molecular , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo , SARS-CoV-2 , Agua/química , Tratamiento Farmacológico de COVID-19
16.
Cell Death Dis ; 11(8): 656, 2020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32814759

RESUMEN

The current epidemic of coronavirus disease-19 (COVID-19) caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) calls for the development of inhibitors of viral replication. Here, we performed a bioinformatic analysis of published and purported SARS-CoV-2 antivirals including imatinib mesylate that we found to suppress SARS-CoV-2 replication on Vero E6 cells and that, according to the published literature on other coronaviruses is likely to act on-target, as a tyrosine kinase inhibitor. We identified a cluster of SARS-CoV-2 antivirals with characteristics of lysosomotropic agents, meaning that they are lipophilic weak bases capable of penetrating into cells. These agents include cepharentine, chloroquine, chlorpromazine, clemastine, cloperastine, emetine, hydroxychloroquine, haloperidol, ML240, PB28, ponatinib, siramesine, and zotatifin (eFT226) all of which are likely to inhibit SARS-CoV-2 replication by non-specific (off-target) effects, meaning that they probably do not act on their 'official' pharmacological targets, but rather interfere with viral replication through non-specific effects on acidophilic organelles including autophagosomes, endosomes, and lysosomes. Imatinib mesylate did not fall into this cluster. In conclusion, we propose a tentative classification of SARS-CoV-2 antivirals into specific (on-target) versus non-specific (off-target) agents based on their physicochemical characteristics.


Asunto(s)
Betacoronavirus/fisiología , Infecciones por Coronavirus/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Neumonía Viral/metabolismo , Replicación Viral/efectos de los fármacos , Animales , Antivirales/farmacología , COVID-19 , Muerte Celular/efectos de los fármacos , Chlorocebus aethiops , Infecciones por Coronavirus/virología , Hidroxicloroquina/farmacología , Mesilato de Imatinib/farmacología , Lisosomas/efectos de los fármacos , Pandemias , Neumonía Viral/virología , Inhibidores de Proteínas Quinasas/farmacología , ARN Viral/efectos de los fármacos , SARS-CoV-2 , Células Vero , Carga Viral/efectos de los fármacos
17.
Clin Pharmacol Ther ; 108(2): 201-211, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32302411

RESUMEN

Azithromycin (AZ) is a broad-spectrum macrolide antibiotic with a long half-life and a large volume of distribution. It is primarily used for the treatment of respiratory, enteric, and genitourinary bacterial infections. AZ is not approved for the treatment of viral infections, and there is no well-controlled, prospective, randomized clinical evidence to support AZ therapy in coronavirus disease 2019 (COVID-19). Nevertheless, there are anecdotal reports that some hospitals have begun to include AZ in combination with hydroxychloroquine or chloroquine (CQ) for treatment of COVID-19. It is essential that the clinical pharmacology (CP) characteristics of AZ be considered in planning and conducting clinical trials of AZ alone or in combination with other agents, to ensure safe study conduct and to increase the probability of achieving definitive answers regarding efficacy of AZ in the treatment of COVID-19. The safety profile of AZ used as an antibacterial agent is well established.1 This work assesses published in vitro and clinical evidence for AZ as an agent with antiviral properties. It also provides basic CP information relevant for planning and initiating COVID-19 clinical studies with AZ, summarizes safety data from healthy volunteer studies, and safety and efficacy data from phase II and phase II/III studies in patients with uncomplicated malaria, including a phase II/III study in pediatric patients following administration of AZ and CQ in combination. This paper may also serve to facilitate the consideration and use of a priori-defined control groups for future research.


Asunto(s)
Antivirales/farmacología , Azitromicina/farmacología , Azitromicina/uso terapéutico , Betacoronavirus/efectos de los fármacos , Antivirales/uso terapéutico , Azitromicina/efectos adversos , Azitromicina/farmacocinética , Betacoronavirus/patogenicidad , Ensayos Clínicos como Asunto , Infecciones por Coronavirus/tratamiento farmacológico , Quimioterapia Combinada , Humanos , Hidroxicloroquina/farmacología , Pulmón/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , SARS-CoV-2 , Tratamiento Farmacológico de COVID-19
19.
Intern Emerg Med ; 12(1): 1-7, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28044251

RESUMEN

The anti-phospholipid syndrome (APS) is an autoimmune disorder characterized by vascular thrombosis and/or pregnancy morbidity, associated with a persistent positivity for anti-phospholipid antibodies (aPL). The current classification criteria for APS include three laboratory tests: lupus anti-coagulant (LA), anti-cardiolipin (aCL), and anti-ß2 glycoprotein-I (ß2GPI). To date, the therapeutic approach for thrombotic APS mainly centers on long-term anti-coagulation with a vitamin K antagonist (VKA). APS management may represent a challenge for the treating physicians. Patients with different aPL profiles need a tailored risk-stratified approach. Moreover, in patients with recurrent thrombotic events despite therapy with VKA, or in those with microvascular involvement, new therapeutic options are highly needed. In this review, we aim to elucidate recent findings about new aPL specifities, available risk scoring models, and novel therapeutic approaches in APS management.


Asunto(s)
Síndrome Antifosfolípido/diagnóstico , Síndrome Antifosfolípido/terapia , Trombosis/terapia , Adulto , Anticuerpos Anticardiolipina/análisis , Anticuerpos Anticardiolipina/sangre , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticoagulantes/farmacología , Anticoagulantes/uso terapéutico , Femenino , Fibrinolíticos/farmacología , Fibrinolíticos/uso terapéutico , Humanos , Hidroxicloroquina/farmacología , Hidroxicloroquina/uso terapéutico , Inhibidor de Coagulación del Lupus/análisis , Inhibidor de Coagulación del Lupus/sangre , Embarazo , Complicaciones del Embarazo/diagnóstico , Complicaciones del Embarazo/prevención & control , Complicaciones del Embarazo/terapia , Medición de Riesgo/métodos , Rivaroxabán/farmacología , Rivaroxabán/uso terapéutico , Trombosis/diagnóstico , beta 2 Glicoproteína I/análisis , beta 2 Glicoproteína I/sangre
20.
Phytomedicine ; 22(10): 902-10, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26321739

RESUMEN

BACKGROUND: Medicinal plants have long been an excellent source of pharmaceutical agents. Autophagy, a catabolic degradation process through lysosomes, plays an important role in tumorigenesis and cancer therapy. PURPOSE: Through a screen designed to identify autophagic regulators from a library of natural compounds, we found that Guttiferone K (GUTK) can activate autophagy in several cancer cell lines. The objective of this study is to investigate the mechanism by which GUTK sensitizes cancer cells to cell death in nutrient starvation condition. METHODS: Cell death analysis was performed by propidium iodide staining with flow cytometry or Annexin V-FITC/PI staining assay. DCFH-DA staining was used for intracellular ROS measurement. Protein levels were analyzed by western blot analysis. Cell viability was measured by MTT assay. RESULTS: Exposure to GUTK was observed to markedly induce GFP-LC3 puncta formation and activate the accumulation of LC3-II and the degradation of p62 in HeLa cells, suggesting that GUTK is an autophagy inducer. Importantly, hydroxychloroquine, an autophagy inhibitor, was found to significantly prevent GUTK-induced cell death in nutrient starvation conditions, suggesting that the cell death observed is largely dependent on autophagy. We further provide evidence that GUTK inhibits Akt phosphorylation, thereby inhibiting the mTOR pathway in cancer cells during nutrient starvation. In addition, GUTK causes the accumulation of reactive oxygen species (ROS) and the phosphorylation of JNK in EBSS, which may mediate both autophagy and apoptosis. CONCLUSION: These data indicate that GUTK sensitizes cancer cells to nutrient stress-induced cell death though Akt/mTOR dependent autophagy pathway.


Asunto(s)
Autofagia , Benzofenonas/farmacología , Antineoplásicos Fitogénicos/farmacología , Muerte Celular , Línea Celular Tumoral , Medios de Cultivo/química , Células HeLa , Humanos , Hidroxicloroquina/farmacología , Proteínas Asociadas a Microtúbulos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas de Unión al ARN/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA