Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Chem Res Toxicol ; 36(7): 1071-1080, 2023 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-37348131

RESUMEN

Overdose of acetaminophen, a widely used antipyretic and analgesic drug, is one of the leading causes of drug-induced acute liver injury in the United States and worldwide. Phase-I metabolism of acetaminophen generates the toxic N-acetyl-p-benzoquinone imine (NAPQI) intermediate. Reactions of NAPQI with a wide range of biomolecules cause increased oxidative stress, endoplasmic reticulum (ER) stress, inflammation, and mitochondrial dysfunction, some of the cellular events contributing toward liver toxicity. Previously, we evaluated the potential of an FDA-approved, ER stress-modulating antihypertensive drug, Wytensin (trans-guanabenz, E-GA), as an antidote for acetaminophen hepatotoxicity. E-GA prevented elevation of the liver enzyme alanine aminotransferase (ALT), even when administered up to 6 h after acetaminophen overdose, and exhibited synergistic analgesic interactions. However, the commercially available guanabenz exists solely as a trans-isomer and suffers from sedative side effects resulting from the inhibition of central α2A-adrenergic receptors in locus coeruleus. Here, we studied the utility of the relatively unexplored cis-isomer of guanabenz as a treatment option for acetaminophen-induced liver toxicity. cis(Z)-Guanabenz acetate (Z-GA) lacks interaction with α2A-adrenoreceptors and is thus devoid of sedative, blood-pressure-lowering side effects of E-GA. Treatment of mice with Z-GA (10 mg/kg) before acetaminophen overdose and up to 6 h post APAP administration prevented liver injury and suppressed the elevation of serum ALT levels. Mechanistically, hepatoprotective effects of both isomers are similar and partly attributed to attenuation of the ER stress and oxidative stress in the liver. The results of this study suggest that Z-GA may be a safer, effective antidote for the clinical management of acute liver injury resulting from acetaminophen overdose. It also raises a tantalizing possibility of a prophylactic combination of the geometric isomer of the approved drug guanabenz with acetaminophen in a clinical setting.


Asunto(s)
Acetaminofén , Enfermedad Hepática Inducida por Sustancias y Drogas , Ratones , Animales , Acetaminofén/toxicidad , Guanabenzo/farmacología , Antídotos/farmacología , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Hígado/metabolismo , Estrés Oxidativo , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo
2.
Optom Vis Sci ; 99(2): 101-113, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34897230

RESUMEN

SIGNIFICANCE: Advances in imaging technology over the last two decades have produced significant innovations in medical imaging. Hyperspectral imaging (HSI) is one of these innovations, enabling powerful new imaging tools for clinical use and greater understanding of tissue optical properties and mechanisms underlying eye disease.Hyperspectral imaging is an important and rapidly growing area in medical imaging, making possible the concurrent collection of spectroscopic and spatial information that is usually obtained from separate optical recordings. In this review, we describe several mainstream techniques used in HSI, along with noteworthy advances in optical technology that enabled modern HSI techniques. Presented also are recent applications of HSI for basic and applied eye research, which include a novel method for assessing dry eye syndrome, clinical slit-lamp examination of corneal injury, measurement of blood oxygen saturation in retinal disease, molecular changes in macular degeneration, and detection of early stages of Alzheimer disease. The review also highlights work resulting from integration of HSI with other imaging tools such as optical coherence tomography and autofluorescence microscopy and discusses the adaptation of HSI for clinical work where eye motion is present. Here, we present the background and main findings from each of these reports along with specific references for additional details.


Asunto(s)
Imágenes Hiperespectrales , Tomografía de Coherencia Óptica , Humanos , Análisis Espectral
3.
Chem Res Toxicol ; 34(5): 1355-1366, 2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-33857375

RESUMEN

Diacetyl (DA), a food flavorant, is linked with occupational lung disease. Our in vitro experiments described the formation of a covalent adduct by DA with Arg5 of the Aß1-42 peptide, which resulted in only a transient increase in neurotoxicity in SH-SY5Y cells. However, in vivo implications of these effects on Alzheimer's disease (AD) pathogenesis and the underlying mechanisms remain poorly understood. In the APP/PS1 transgenic AD mouse model, DA treatment did not exacerbate learning and memory deficits in the Morris water maze test. Moreover, DA increased the Aß1-42 plaque burden and decreased neuronal inflammation in the transgenic AD mice. Additionally, cognitive impairment induced by intracerebroventricular Aß1-42 was restored by the DA treatment, as assessed by the T-maze test. A corresponding mitigation of neuronal inflammation was also observed in the hippocampus of these nontransgenic mice due to the acceleration of Aß1-42 aggregation by DA into nontoxic plaques. The data from SDS-PAGE, dot-blot, and TEM in vitro experiments corroborated the acceleration of the Aß1-42 aggregation observed in vivo in AD animal models and characterized the DA-induced formation of Aß1-42 fibrils. Such Aß1-42-DA fibrils were unstable in the presence of detergent and amenable to detection by the thioflavin T reagent, thus underscoring the distinct assembly of these fibrils compared to that of the fibrils of the native Aß1-42. Taken together, the results of this study present for the first time the in vivo implications of the DA-induced acceleration of Aß1-42 and may provide a strategy for the rational design of Aß1-42 aggregation accelerators as AD therapeutics that promote oligomer-free Aß1-42 fibril formation.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/antagonistas & inhibidores , Disfunción Cognitiva/tratamiento farmacológico , Diacetil/farmacología , Modelos Animales de Enfermedad , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/antagonistas & inhibidores , Administración Oral , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Disfunción Cognitiva/metabolismo , Diacetil/administración & dosificación , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Fragmentos de Péptidos/metabolismo , Agregado de Proteínas/efectos de los fármacos , Células Tumorales Cultivadas
4.
Chem Res Toxicol ; 33(1): 162-171, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31524377

RESUMEN

Endoplasmic reticulum (ER) stress has been shown to be involved in the hepatotoxicity of acetaminophen (APAP). Guanabenz (GA), a widely known antihypertensive drug, is reported to exhibit an anti-ER stress effect. In this study, we investigated the potential of GA as an antidote against APAP-induced hepatotoxicity. The underlying biochemical mechanisms for the hepatoprotective effect of GA were explored. Here we found that treatment of mice with GA (10 mg/kg) before APAP overdose dramatically prevented APAP-induced liver enzyme elevation and resultant toxicity in mice, as indicated by suppression of elevated serum alanine aminotransferase (ALT) levels and liver histological analysis. Importantly, delayed administration of GA within 6 h after APAP overdose also showed an almost equivalent protective effect against APAP liver toxicity. Mechanistically, several pathways are involved in the protective effect of GA against APAP-induced live toxicity, including attenuation of ER stress and oxidative stress, increased levels of nontoxic phase I and II metabolites of APAP, decrease in the formation of toxic N-acetyl-p-benzoquinone imine (NAPQI), and its subsequent protein binding. Importantly, combination of GA with APAP exhibited synergistic interaction in the latter's analgesic activity, while sparing its antipyretic action. These findings provide the preclinical evidence of GA as a promising antidote for treatment of APAP-induced liver toxicity and raise a possibility of its combination with APAP in clinical settings.


Asunto(s)
Acetaminofén , Analgésicos no Narcóticos/uso terapéutico , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Guanabenzo/uso terapéutico , Sustancias Protectoras/uso terapéutico , Alanina Transaminasa/sangre , Analgesia , Analgésicos no Narcóticos/farmacología , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Sinergismo Farmacológico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Glutatión/metabolismo , Guanabenzo/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Estrés Oxidativo/efectos de los fármacos , Sustancias Protectoras/farmacología
5.
Chem Res Toxicol ; 30(3): 777-784, 2017 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-28165728

RESUMEN

Ψ-Glutathione (ψ-GSH) is an orally bioavailable and metabolism-resistant glutathione analogue that has been shown previously to substitute glutathione in most of its biochemical roles. Described here in its entirety is the preclinical evaluation of ψ-GSH as a rescue agent for acetaminophen (APAP) overdose: an event where time is of essence. By employing a murine model, four scenarios commonly encountered in emergency medicine are reconstructed. ψ-GSH is juxtaposed against N-acetylcysteine (NAC), the sole clinically available drug, in each of the scenarios. While both agents appear to be equally efficacious when timely administered, ψ-GSH partly retains its efficacy even in the face of substantial delay in administration. Thus, implied is the ability of ψ-GSH to intercept secondary toxicology following APAP insult. Oral availability and complete lack of toxicity as evaluated by liver function tests and survival analysis underscored ψ-GSH as a safer and more efficacious alternative to NAC. Finally, the pharmacodynamic mimicry of GSH by ψ-GSH is illustrated through the isolation and chemical characterization of an entity that can arise only through direct encounter of ψ-GSH with N-acetyl-p-benzoquinoneimine, the primary toxic metabolite of APAP.


Asunto(s)
Acetaminofén/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Modelos Animales de Enfermedad , Glutatión/farmacología , Animales , Ratones
6.
Bioorg Med Chem Lett ; 24(1): 332-6, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24269162

RESUMEN

Three classes of novel inhibitors of inosine monophosphate dehydrogenase have been prepared and their anti-proliferative properties were evaluated against several cancer cell lines. (1) Mycophenolic adenine dinucleotide analogues (8-13) containing a substituent at the C2 of adenine ring were found to be potent inhibitors of IMPDH (Ki's in range of 0.6-82nM) and sub-µM inhibitors of leukemic K562 cell proliferation. (2) Mycophenolic adenosine (d and l) esters (20 and 21) showed a potent inhibition of IMPDH2 (Ki=102 and Ki=231nM, respectively) and inhibition of K562 cell growth (IC50=0.5 and IC50=1.6µM). These compounds serve both as inhibitors of the enzyme and as a depot form of mycophenolic acid. The corresponding amide analogue 22, also a potent inhibitor of IMPDH (Ki=84nM), did not inhibit cancer cell proliferation. (3) Mycophenolic-(l)- and (d)-valine adenine di-amide derivatives 25 (Ki=9nM) and 28 (Ki=3nM) were found to be very potent enzymatically, but did not inhibit proliferation of cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , NAD/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Células HT29 , Células HeLa , Humanos , IMP Deshidrogenasa/antagonistas & inhibidores , IMP Deshidrogenasa/metabolismo , Células K562 , Modelos Moleculares , Estructura Molecular , NAD/análogos & derivados , NAD/química , Relación Estructura-Actividad
7.
Sci Rep ; 14(1): 13838, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38879582

RESUMEN

According to the Intergovernmental Panel on Climate Change (IPCC) of the United Nations (UN), rise in atmospheric concentration of carbon dioxide (CO 2 ) due to anthropogenic factors is considered as the primary driver for global climate change. With almost every major corporation around the world working towards their "net-zero goals", it is becoming increasingly important to have more technologies that can help reduce carbon footprint. Achieving sequestration of CO 2 in the subsurface through Carbon Capture Utilization and Storage (CCUS) technologies like CO 2 -Enhanced Oil Recovery, CO 2 -Enhanced Geothermal Systems, CO 2 -Enhanced Coal Bed Methane, etc. is well accepted. We introduce yet another attractive CCUS opportunity through well rehabilitation. Aqua Freed® and Aqua Gard® are well-known well rehabilitation and preventive well maintenance technologies that utilize (inject underground) liquid CO 2 for the purpose. The goal of this study was to quantify the storage capacity of Aqua Freed® and Aqua Gard®, and establish their CCUS credentials. Depending on the well being serviced, these technologies can inject up to 40 US tons of CO 2 per well. Based on field data collection and statistical modeling, we estimated that 82-96% (median 90%) of the injected CO 2 remains in the subsurface post injection. Overall, our results and analysis of the US market suggest that using CO 2 for well rehabilitation and maintenance has a storage potential of several megatonnes of CO 2 annually in the US alone.

8.
Eur J Med Chem ; 275: 116604, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38917665

RESUMEN

The endogenous opioid system regulates pain through local release of neuropeptides and modulation of their action on opioid receptors. However, the effect of opioid peptides, the enkephalins, is short-lived due to their rapid hydrolysis by enkephalin-degrading enzymes. In turn, an innovative approach to the management of pain would be to increase the local concentration and prolong the stability of enkephalins by preventing their inactivation by neural enkephalinases such as puromycin-sensitive aminopeptidase (PSA). Our previous structure-activity relationship studies offered the S-diphenylmethyl cysteinyl derivative of puromycin (20) as a nanomolar inhibitor of PSA. This chemical class, however, suffered from undesirable metabolism to nephrotoxic puromycin aminonucleoside (PAN). To prevent such toxicity, we designed and synthesized 5'-chloro substituted derivatives. The compounds retained the PSA inhibitory potency of the corresponding 5'-hydroxy analogs and had improved selectivity toward PSA. In vivo treatment with the lead compound 19 caused significantly reduced pain response in antinociception assays, alone and in combination with Met-enkephalin. The analgesic effect was reversed by the opioid antagonist naloxone, suggesting the involvement of opioid receptors. Further, PSA inhibition by compound 19 in brain slices caused local increase in endogenous enkephalin levels, corroborating our rationale. Pharmacokinetic assessment of compound 19 showed desirable plasma stability and identified the cysteinyl sulfur as the principal site of metabolic liability. We gained additional insight into inhibitor-PSA interactions by molecular modeling, which underscored the importance of bulky aromatic amino acid in puromycin scaffold. The results of this study strongly support our rationale for the development of PSA inhibitors for effective pain management.

9.
Antioxidants (Basel) ; 13(6)2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38929087

RESUMEN

Acetaminophen (APAP) overdose triggers a cascade of intracellular oxidative stress events, culminating in acute liver injury. The clinically used antidote, N-acetylcysteine (NAC), has a narrow therapeutic window, and early treatment is essential for a satisfactory therapeutic outcome. For more versatile therapies that can be effective even at late presentation, the intricacies of APAP-induced hepatotoxicity must be better understood. Accumulation of advanced glycation end products (AGEs) and the consequent activation of the receptor for AGEs (RAGE) are considered one of the key mechanistic features of APAP toxicity. Glyoxalase 1 (Glo-1) regulates AGE formation by limiting the levels of methylglyoxal (MEG). In this study, we studied the relevance of Glo-1 in the APAP-mediated activation of RAGE and downstream cell death cascades. Constitutive Glo-1-knockout mice (GKO) and a cofactor of Glo-1, ψ-GSH, were used as tools. Our findings showed elevated oxidative stress resulting from the activation of RAGE and hepatocyte necrosis through steatosis in GKO mice treated with high-dose APAP compared to wild-type controls. A unique feature of the hepatic necrosis in GKO mice was the appearance of microvesicular steatosis as a result of centrilobular necrosis, rather than the inflammation seen in the wild type. The GSH surrogate and general antioxidant ψ-GSH alleviated APAP toxicity irrespective of the Glo-1 status, suggesting that oxidative stress is the primary driver of APAP toxicity. Overall, the exacerbation of APAP hepatotoxicity in GKO mice suggests the importance of this enzyme system in antioxidant defense against the initial stages of APAP overdose.

10.
bioRxiv ; 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38617237

RESUMEN

The endogenous opioid system regulates pain through local release of neuropeptides and modulation of their action on opioid receptors. However, the effect of opioid peptides, the enkephalins, is short-lived due to their rapid hydrolysis by enkephalin-degrading enzymes. In turn, an innovative approach to the management of pain would be to increase the local concentration and prolong the stability of enkephalins by preventing their inactivation by neural enkephalinases such as puromycin sensitive aminopeptidase (PSA). Our previous structure-activity relationship studies offered the S-diphenylmethyl cysteinyl derivative of puromycin (20) as a nanomolar inhibitor of PSA. This chemical class, however, suffered from undesirable metabolism to nephrotoxic puromycin aminonucleoside (PAN). To prevent such toxicity, we designed and synthesized 5'-chloro substituted derivatives. The compounds retained the PSA inhibitory potency of the corresponding 5'-hydroxy analogs and had improved selectivity toward PSA. In vivo treatment with the lead compound 19 caused significantly reduced pain response in antinociception assays, alone and in combination with Met-enkephalin. The analgesic effect was reversed by the opioid antagonist naloxone, suggesting the involvement of opioid receptors. Further, PSA inhibition by compound 19 in brain slices caused local increase in endogenous enkephalin levels, corroborating our rationale. Pharmacokinetic assessment of compound 19 showed desirable plasma stability and identified the cysteinyl sulfur as the principal site of metabolic liability. We gained additional insight into inhibitor-PSA interactions by molecular modeling, which underscored the importance of bulky aromatic amino acid in puromycin scaffold. The results of this study strongly support our rationale for the development of PSA inhibitors for effective pain management.

11.
Bioorg Med Chem Lett ; 23(3): 620-3, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23290453

RESUMEN

Excessive exposure to sunlight is primarily implicated in ultraviolet (UV) induced skin cancers worldwide. Direct absorption of UV radiation by DNA leads to the formation of cyclobutane pyrimidine dimers (CPDs) resulting in DNA damage. The molecular mechanisms involved in the mutagenicity of CPDs are well established. Photoprotection of the skin from the detrimental effects of UV is essential in preventing skin damage. A variety of formulations, which essentially contain UV filters have been used as photoprotective agents of the skin. These comprise aromatic and inorganic molecules, whose mechanism of action involves either absorption, reflection, or scattering of UV radiation. However, the downstream photoproducts of some of these molecules have undesirable characteristics which compromise their utility. A biomimetic approach involving structural analogs of nucleic acids can help overcome these limitations. Herein, we show the photoprotective action of acyclothymidine dinucleosides on both plasmid and cellular DNA.


Asunto(s)
ADN/efectos de los fármacos , Fosfatos de Dinucleósidos/química , Protectores Solares/química , Uracilo/análogos & derivados , ADN/efectos de la radiación , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Fosfatos de Dinucleósidos/farmacología , Electroforesis en Gel de Agar , Plásmidos/efectos de los fármacos , Plásmidos/efectos de la radiación , Protectores Solares/farmacología , Uracilo/química , Uracilo/farmacología , Agua/química
12.
bioRxiv ; 2023 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-38187538

RESUMEN

Acetaminophen (APAP) overdose triggers a cascade of intracellular oxidative stress events culminating in acute liver injury. The clinically used antidote, N-acetylcysteine (NAC) has a narrow therapeutic window and early treatment is essential for satisfactory therapeutic outcome. For more versatile therapies that can be effective even at late-presentation, the intricacies of APAP-induced hepatotoxicity must be better understood. Accumulation of advanced glycation end-products (AGEs) and consequent activation of the receptor for AGEs (RAGE) are considered one of the key mechanistic features of APAP toxicity. Glyoxalase-1 (Glo-1) regulates AGE formation by limiting the levels of methylglyoxal (MEG). In this study, we studied the relevance of Glo-1 in APAP mediated activation of RAGE and downstream cell-death cascades. Constitutive Glo-1 knockout mice (GKO) and a cofactor of Glo-1, ψ-GSH, were employed as tools. Our findings show elevated oxidative stress, activation of RAGE and hepatocyte necrosis through steatosis in GKO mice treated with high-dose APAP compared to wild type controls. A unique feature of the hepatic necrosis in GKO mice is the appearance of microvesicular steatosis as a result of centrilobular necrosis, rather than inflammation seen in wild type. The GSH surrogate and general antioxidant, ψ-GSH alleviated APAP toxicity irrespective of Glo-1 status, suggesting that oxidative stress being the primary driver of APAP toxicity. Overall, exacerbation of APAP hepatotoxicity in GKO mice suggests the importance of this enzyme system in antioxidant defense against initial stages of APAP overdose.

13.
Chem Res Toxicol ; 25(10): 2083-91, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22731744

RESUMEN

Diacetyl (DA), an ubiquitous butter-flavoring agent, was found to influence several aspects of amyloid-ß (Aß) aggregation--one of the two primary pathologies associated with Alzheimer's disease. Thioflavin T fluorescence and circular dichroism spectroscopic measurements revealed that DA accelerates A߹⁻4² aggregation into soluble and ultimately insoluble ß-pleated sheet structures. DA was found to covalently bind to Arg5 of A߹⁻4² through proteolytic digestion-mass spectrometric experiments. These biophysical and chemical effects translated into the potentiation of A߹⁻4² cytotoxicity by DA toward SH-SY5Y cells in culture. DA easily traversed through a MDR1-MDCK cell monolayer, an in vitro model of the blood-brain barrier. Additionally, DA was found not only to be resistant to but also inhibitory toward glyoxalase I, the primary initiator of detoxification of amyloid-promoting reactive dicarbonyl species that are generated naturally in large amounts by neuronal tissue. In light of the chronic exposure of industry workers to DA, this study raises the troubling possibility of long-term neurological toxicity mediated by DA.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Mantequilla/toxicidad , Diacetil/toxicidad , Aromatizantes/toxicidad , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/química , Animales , Benzotiazoles , Barrera Hematoencefálica/metabolismo , Línea Celular , Supervivencia Celular , Dicroismo Circular , Diacetil/metabolismo , Perros , Aromatizantes/metabolismo , Colorantes Fluorescentes/análisis , Colorantes Fluorescentes/metabolismo , Humanos , Lactoilglutatión Liasa/metabolismo , Células de Riñón Canino Madin Darby , Fragmentos de Péptidos/química , Permeabilidad , Conformación Proteica/efectos de los fármacos , Tiazoles/análisis , Tiazoles/metabolismo
14.
Bioorg Med Chem ; 20(15): 4790-800, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22748708

RESUMEN

Quinolone-3-carboxylic acid represents a highly privileged chemotype in medicinal chemistry and has been extensively explored as antibiotics and antivirals targeting human immunodeficiency virus (HIV) integrase (IN). Herein we describe the synthesis and anti-hepatitis C virus (HCV) profile of a series of C-6 aryl substituted 4-quinlone-3-carboxylic acid analogues. Significant inhibition was observed with a few analogues at low micromolar range against HCV replicon in cell culture and a reduction in replicon RNA was confirmed through an RT-qPCR assay. Interestingly, evaluation of analogues as inhibitors of NS5B in a biochemical assay yielded only modest inhibitory activities, suggesting that a different mechanism of action could operate in cell culture.


Asunto(s)
4-Quinolonas/farmacología , Antivirales/farmacología , Ácidos Carboxílicos/farmacología , Inhibidores Enzimáticos/farmacología , Hepacivirus/efectos de los fármacos , Replicón/efectos de los fármacos , Proteínas no Estructurales Virales/antagonistas & inhibidores , 4-Quinolonas/síntesis química , 4-Quinolonas/química , Antivirales/síntesis química , Antivirales/química , Ácidos Carboxílicos/síntesis química , Ácidos Carboxílicos/química , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Hepacivirus/genética , Humanos , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Reacción en Cadena en Tiempo Real de la Polimerasa , Replicón/genética , Relación Estructura-Actividad , Proteínas no Estructurales Virales/metabolismo
15.
Bioorg Med Chem ; 20(1): 467-79, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22100256

RESUMEN

C7-Substituted 2-hydroxyisoquinoline-1,3-diones inhibit the strand transfer of HIV integrase (IN) and the reverse-transcriptase-associated ribonuclease H (RNH). Hepatitis C virus (HCV) NS5B polymerase shares a similar active site fold to RNH and IN, suggesting that N-hydroxyimides could be useful inhibitor scaffolds of HCV via targeting the NS5B. Herein we describe the design, chemical synthesis, replicon and biochemical assays, and molecular docking of C-6 or C-7 aryl substituted 2-hydroxyisoquinoline-1,3-diones as novel HCV inhibitors. The synthesis involved an improved and clean cyclization method, which allowed the convenient preparation of various analogs. Biological studies revealed that the C-6 analogs, a previously unknown chemotype, consistently inhibit both HCV replicon and recombinant NS5B at low micromolar range. Molecular modeling studies suggest that these inhibitors may bind to the NS5B active site.


Asunto(s)
Antivirales/síntesis química , Antivirales/farmacología , Hepacivirus/efectos de los fármacos , Isoquinolinas/química , Antivirales/química , Sitios de Unión , Dominio Catalítico , Simulación por Computador , Ciclización , Diseño de Fármacos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Hepacivirus/enzimología , Isoquinolinas/síntesis química , Isoquinolinas/farmacología , Relación Estructura-Actividad , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/efectos de los fármacos
16.
Anesth Analg ; 114(5): 956-61, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22392971

RESUMEN

BACKGROUND: Cyanide (CN) toxicity is a serious clinical problem and can occur with sodium nitroprusside (SNP) administration, accidental smoke inhalation, industrial mishaps, and bio-terrorism. In this study, we induced severe CN toxicity independently with SNP or sodium cyanide (NaCN) in a juvenile pig model to demonstrate reversal of severe CN toxicity with a new antidote, sulfanegen sodium, a prodrug of 3-mercaptopyruvate. METHODS: SNP study: A pilot study in 11 anesthetized, mechanically ventilated juvenile pigs allowed us to determine the dose of SNP to induce CN toxicity. Blood CN, serum lactates, and blood gases were monitored. CN toxicity was defined as the occurrence of severe lactic acidosis accompanied by significant elevation in blood CN levels. Based on this pilot study, 8 anesthetized pigs received a high-dose i.v. infusion of SNP (100 mg/h) for 2 hours to induce CN toxicity. They were then randomized to receive either sulfanegen sodium or placebo. Four pigs received 3 doses of sulfanegen sodium (2.5 g i.v.) every hour after induction of severe CN toxicity, and 4 pigs received placebo. NaCN study: A pilot study was conducted in 4 spontaneously ventilating pigs sedated with propofol plus ketamine to demonstrate hemodynamic and metabolic stability for several hours. After this, 6 pigs were similarly sedated and given NaCN in bolus aliquots to produce CN toxicity ultimately resulting in death. Hemodynamics and metabolic variables were followed to define peak CN toxicity. In another group of 6 pigs, severe CN toxicity was induced by this method, and at peak toxicity, the animals were given sulfanegen sodium (2.5 g i.v.) followed by a repeat dose 60 minutes later in surviving animals. RESULTS: SNP study: The pilot study demonstrated the occurrence of a significant increase in blood CN levels (P < 0.05) accompanied by severe lactic acidemia (P < 0.05) in all pigs receiving a high dose of SNP. Administration of the sulfanegen antidote resulted in progressive significant reduction in blood lactate and CN levels with 100% survival (P < 0.05), whereas the placebo-treated pigs deteriorated and did not survive (P < 0.05). NaCN study: NaCN injection resulted in CN toxicity accompanied by severe lactic acidosis and mortality in all the pigs. Sulfanegen sodium reversed this toxicity and prevented mortality in all the pigs treated with this antidote. CONCLUSIONS: CN toxicity can be successfully induced in a juvenile pig model with SNP or NaCN. The prodrug, sulfanegen sodium, is effective in reversing CN toxicity induced by SNP or NaCN.


Asunto(s)
Cianuros/antagonistas & inhibidores , Cianuros/toxicidad , Cisteína/análogos & derivados , Compuestos Heterocíclicos con 1 Anillo/farmacología , Profármacos/farmacología , Animales , Análisis de los Gases de la Sangre , Presión Sanguínea/efectos de los fármacos , Presión Venosa Central/efectos de los fármacos , Cianuros/sangre , Cisteína/farmacología , Frecuencia Cardíaca/efectos de los fármacos , Concentración de Iones de Hidrógeno , Ácido Láctico/sangre , Nitroprusiato/efectos adversos , Proyectos Piloto , Arteria Pulmonar/efectos de los fármacos , Porcinos , Vasodilatadores/efectos adversos
17.
Antioxidants (Basel) ; 11(6)2022 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-35739972

RESUMEN

Supplementation of glutathione (GSH) levels through varying formulations or precursors has thus far appeared to be a tenable strategy to ameliorate disease-associated oxidative stress. Metabolic liability of GSH and its precursors, i.e., hydrolysis by the ubiquitous γ-glutamyl transpeptidase (γ-GT), has limited successful clinical translation due to poor bioavailability. We addressed this problem through the design of γ-GT-resistant GSH analogue, ψ-GSH, which successfully substituted in GSH-dependent enzymatic systems and also offered promise as a therapeutic for Alzheimer's disease (AD). With the aim to improve its bioavailability, we studied the utility of a ψ-GSH precursor, dipeptide 2, as a potential AD therapeutic. Compound 2 retains the γ-GT stable ureide linkage and the thiol group for antioxidant property. By engaging glutathione synthetase, compound 2 was able to generate ψ-GSH in vivo. It was found to be a modest cofactor of glutathione peroxidase and prevented cytotoxicity of Aß1-42-aggregates in vitro. Studies of compound 2 in an acute AD model generated by intracerebroventricular injection of Aß1-42 showed cognitive benefits, which were augmented by its combination with glycine along with mitigation of oxidative stress and inflammatory pathology. Collectively, these results support further optimization and evaluation of ψ-GSH dipeptide as a potential therapeutic in transgenic AD models.

18.
J Med Chem ; 65(21): 14441-14455, 2022 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-36353871

RESUMEN

Addressing glycation-induced oxidative stress in Alzheimer's disease (AD) is an emerging pharmacotherapeutic strategy. Restoration of the brain glyoxalase enzyme system that neutralizes reactive dicarbonyls is one such approach. Toward this end, we designed, synthesized, and evaluated a γ-glutamyl transpeptidase-resistant glyoxalase substrate, ψ-GSH. Although mechanistically successful, the oral efficacy of ψ-GSH appeared as an area in need of improvement. Herein, we describe our rationale for the creation of prodrugs that mask the labile sulfhydryl group. In vitro and in vivo stability studies identified promising prodrugs that could deliver pharmacologically relevant brain levels of ψ-GSH. When administered orally to a mouse model generated by the intracerebroventricular injection of Aß1-42, the compounds conferred cognitive benefits. Biochemical and histological examination confirmed their effects on neuroinflammation and oxidative stress. Collectively, we have identified orally efficacious prodrugs of ψ-GSH that are able to restore brain glyoxalase activity and mitigate inflammatory and oxidative pathology associated with AD.


Asunto(s)
Enfermedad de Alzheimer , Lactoilglutatión Liasa , Profármacos , Animales , Ratones , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Profármacos/farmacología , Profármacos/uso terapéutico , Estrés Oxidativo , Modelos Animales de Enfermedad , Péptidos beta-Amiloides/farmacología
19.
Redox Biol ; 57: 102484, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36183541

RESUMEN

Increased oxidative stress and inflammation are implicated in the pathogenesis of Alzheimer's disease. Treatment with hydrogen sulfide (H2S) and H2S donors such as sodium hydrosulfide (NaSH) can reduce oxidative stress in preclinical studies, however clinical benefits of such treatments are rather ambiguous. This is partly due to poor stability and bioavailability of the H2S donors, requiring impractically large doses that are associated with dose-limiting toxicity. Herein, we identified a bioavailable 3-mercaptopyruvate prodrug, sulfanegen, which is able to pose as a sacrificial redox substrate for 3-mercaptopyruvate sulfurtransferase (3MST), one of the H2S biosynthetic enzymes in the brain. Sulfanegen is able to mitigate toxicity emanating from oxidative insults and the Aß1-42 peptide by releasing H2S through the 3MST pathway. When administered to symptomatic transgenic mouse model of AD (APP/PS1; 7 and 12 months) and mice that were intracerebroventricularly administered with the Aß1-42 peptide, sulfanegen was able to reverse oxidative and neuroinflammatory consequences of AD pathology by restoring 3MST function. Quantitative neuropathological analyses confirmed significant disease modifying effect of the compound on amyloid plaque burden and brain inflammatory markers. More importantly, sulfanegen treatment attenuated progressive neurodegeneration in these mice, as evident from the restoration of TH+ neurons in the locus coeruleus. This study demonstrates a previously unknown concept that supplementation of 3MST function in the brain may be a viable approach for the management of AD. Finally, brought into the spotlight is the potential of sulfanegen as a promising AD therapeutic for future drug development efforts.

20.
Bioorg Med Chem Lett ; 21(8): 2400-2, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21392991

RESUMEN

N-3-hydroxylation of pyrimidine-2,4-diones was recently found to yield inhibitors of both HIV-1 reverse transcriptase (RT) and integrase (IN). An extended series of analogues featuring a benzoyl group at the C-6 position of the pyrimidine ring was synthesized. Through biochemical studies it was found that these new analogues are dually active against both RT and IN in low micromolar range. Antiviral assays confirmed that these new inhibitors are active against HIV-1 in cell culture at nanomolar to low micromolar range, further validating 3-hydroxypyrimidine-2,4-diones as a viable scaffold for antiviral development.


Asunto(s)
Inhibidores de Integrasa VIH/química , Integrasa de VIH/química , Transcriptasa Inversa del VIH/química , VIH-1/enzimología , Pirimidinas/química , Inhibidores de la Transcriptasa Inversa/química , Sitios de Unión , Simulación por Computador , Integrasa de VIH/metabolismo , Inhibidores de Integrasa VIH/síntesis química , Inhibidores de Integrasa VIH/farmacología , Transcriptasa Inversa del VIH/metabolismo , VIH-1/efectos de los fármacos , Humanos , Pirimidinas/síntesis química , Pirimidinas/farmacología , Inhibidores de la Transcriptasa Inversa/síntesis química , Inhibidores de la Transcriptasa Inversa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA