Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
bioRxiv ; 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38617237

ABSTRACT

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.

2.
Chem Res Toxicol ; 36(7): 1071-1080, 2023 07 17.
Article in English | MEDLINE | ID: mdl-37348131

ABSTRACT

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.


Subject(s)
Acetaminophen , Chemical and Drug Induced Liver Injury , Mice , Animals , Acetaminophen/toxicity , Guanabenz/pharmacology , Antidotes/pharmacology , Disease Models, Animal , Mice, Inbred C57BL , Liver/metabolism , Oxidative Stress , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/prevention & control , Chemical and Drug Induced Liver Injury/metabolism
3.
bioRxiv ; 2023 Dec 23.
Article in English | MEDLINE | ID: mdl-38187538

ABSTRACT

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.

4.
J Med Chem ; 65(21): 14441-14455, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36353871

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Lactoylglutathione Lyase , Prodrugs , Animals , Mice , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Prodrugs/pharmacology , Prodrugs/therapeutic use , Oxidative Stress , Disease Models, Animal , Amyloid beta-Peptides/pharmacology
5.
Redox Biol ; 57: 102484, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36183541

ABSTRACT

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.

6.
Antioxidants (Basel) ; 11(6)2022 May 28.
Article in English | MEDLINE | ID: mdl-35739972

ABSTRACT

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.

7.
Optom Vis Sci ; 99(2): 101-113, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34897230

ABSTRACT

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.


Subject(s)
Hyperspectral Imaging , Tomography, Optical Coherence , Humans , Spectrum Analysis
8.
Antioxidants (Basel) ; 10(11)2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34829667

ABSTRACT

Oxidative stress in Alzheimer's disease (AD) is mediated, in part, by the loss of glutathione (GSH). Previous studies show that γ-glutamyl transpeptidase (GGT)-resistant GSH analog, Ψ-GSH, improves brain GSH levels, reduces oxidative stress markers in brains of APP/PS1 transgenic mice, a mouse model of AD, and attenuates early memory deficits in the APP/PS1 model. Herein, we examined whether Ψ-GSH can attenuate the disease progression when administered following the onset of AD-like pathology in vivo. Cohorts of APP/PS1 mice were administered Ψ-GSH for 2 months starting at 8 month or 12 months of age. We show that Ψ-GSH treatment reduces indices of oxidative stress in older mice by restoration of enzyme glyoxalase-1 (Glo-1) activity and reduces levels of insoluble Aß. Quantitative neuropathological analyses show that Ψ-GSH treatment significantly reduces Aß deposition and brain inflammation in APP/PS1 mice compared to vehicle-treated mice. More importantly, Ψ-GSH treatment attenuated the progressive loss of cortical TH+ afferents and the loss of TH+ neurons in the locus coeruleus (LC). Collectively, the results show that Ψ-GSH exhibits significant antioxidant activity in aged APP/PS1 mice and chronic Ψ-GSH treatment administered after the onset of AD pathology can reverse/slow further progression of AD-like pathology and neurodegeneration in vivo.

9.
Antioxidants (Basel) ; 10(6)2021 Jun 10.
Article in English | MEDLINE | ID: mdl-34200599

ABSTRACT

L-Cysteine-glutathione mixed disulfide (CySSG), a prodrug of glutathione (GSH), was found to be orally bioavailable in mice, and protected mice against a toxic dose of acetaminophen. If oral bioavailability can also be demonstrated in humans, a wide range of applicability for CySSG can be envisioned.

10.
Chem Res Toxicol ; 34(5): 1355-1366, 2021 05 17.
Article in English | MEDLINE | ID: mdl-33857375

ABSTRACT

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.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Cognitive Dysfunction/drug therapy , Diacetyl/pharmacology , Disease Models, Animal , Neuroprotective Agents/pharmacology , Peptide Fragments/antagonists & inhibitors , Administration, Oral , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Cognitive Dysfunction/metabolism , Diacetyl/administration & dosage , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Neuroprotective Agents/administration & dosage , Peptide Fragments/metabolism , Protein Aggregates/drug effects , Tumor Cells, Cultured
11.
Chem Res Toxicol ; 33(1): 162-171, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31524377

ABSTRACT

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.


Subject(s)
Acetaminophen , Analgesics, Non-Narcotic/therapeutic use , Chemical and Drug Induced Liver Injury/drug therapy , Guanabenz/therapeutic use , Protective Agents/therapeutic use , Alanine Transaminase/blood , Analgesia , Analgesics, Non-Narcotic/pharmacology , Animals , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Drug Synergism , Endoplasmic Reticulum Stress/drug effects , Glutathione/metabolism , Guanabenz/pharmacology , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Oxidative Stress/drug effects , Protective Agents/pharmacology
12.
ACS Chem Neurosci ; 10(11): 4492-4501, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31603648

ABSTRACT

A noninvasive and cost-effective means to detect preclinical Alzheimer's disease (AD) and monitor disease progression would be invaluable. The retina is a developmental extension of the brain and has been viewed as a window to evaluate AD-related pathology. Cross-sectional studies have shown structural changes in the retina of AD patients that include thinning of the retinal nerve-fiber layer and changes in retinal vasculature. However, such changes do not manifest in early stages of the disease nor are they specific biomarkers for AD. Described herein is the utilization of our retinal hyperspectral imaging (rHSI) technique as a biomarker for identification of AD-related early pathological changes in the retina. Specifically, this account concerns the translation of our rHSI technique from animal models to human AD subjects. The underlying principle is Rayleigh light scattering, which is expected from low-order Aß aggregates present in early pathology. Recruitment was restricted to AD subjects (N = 19) and age-matched controls, with no family history of AD (N = 16). To limit the influence of skin pigmentation, subjects were restricted to those with skin pigmentation values of 2-3 on the Fitzpatrick scale. The largest spectral deviation from control subjects, rHSI signature, was obtained at the MCI stage with MMSE scores ⩾22, suggesting higher sensitivity of this technique in early disease stages. The rHSI signature observed is unaffected by eye pathologies such as glaucoma and cataract. Age of the subjects minimally influenced the spectral signatures. The rHSI technique shows promise for detection of preclinical AD; it is conducted in a truly noninvasive manner, without application of an exogenous label, and is thus potentially suitable for population screening.


Subject(s)
Alzheimer Disease/diagnostic imaging , Image Processing, Computer-Assisted/methods , Optical Imaging/methods , Retina/diagnostic imaging , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Biomarkers/metabolism , Cohort Studies , Early Diagnosis , Female , Humans , Male , Middle Aged , Retina/metabolism
13.
ACS Chem Neurosci ; 10(9): 4007-4017, 2019 09 18.
Article in English | MEDLINE | ID: mdl-31415151

ABSTRACT

Tosedostat is an inhibitor of aminopeptidases currently in phase II clinical trials for the treatment of blood-related cancers. In our laboratories, we have discovered that it possesses analgesic properties. Extensive in vivo pharmacological studies for the determination of antinociceptive effects of tosedostat are presented here. These studies have indicated that the observed analgesic effect of tosedostat stems from its action on the peripheral nervous system with minimal contribution from the central nervous system. Additionally, when given in combination with morphine, tosedostat exerts a synergistic analgesic effect resulting in a reduction of effective dosages required to achieve the same analgesic effect. With broad implications in addressing the opioid addiction crisis, these revelations attest to tosedostat being a highly valuable drug candidate with diverse pharmacological functions.


Subject(s)
Analgesics/pharmacology , Antineoplastic Agents/pharmacology , Drug Discovery/methods , Glycine/analogs & derivatives , Hydroxamic Acids/pharmacology , Pain Measurement/drug effects , Pain Measurement/methods , Analgesics/chemistry , Animals , Antineoplastic Agents/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Glycine/chemistry , Glycine/pharmacology , Hydroxamic Acids/chemistry , Male , Mice , Molecular Docking Simulation/methods , Narcotic Antagonists/pharmacology
14.
Eur J Med Chem ; 139: 325-336, 2017 Oct 20.
Article in English | MEDLINE | ID: mdl-28803047

ABSTRACT

Substantial progress has been described in the study of puromycin and its analogs for antibiotic properties. However, the peptidase inhibitory activity of related analogs has not been explored as extensively. Specifically, inhibiting aminopeptidases for achieving antitumor effect has been sparsely investigated. Herein, we address this challenge by reporting the synthesis of a series of analogs based on the structural template of puromycin. We also present exhaustive biochemical and in vitro analyses in support of our thesis. Analyzing the structure-activity relationship revealed a steric requirement for maximum potency. Effective inhibitors of Puromycin-Sensitive Aminopeptidase (PSA) are disclosed here. These potential therapeutic agents display superior in vitro antitumor potency against two leukemic cell lines, as compared to known inhibitors of aminopeptidases.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Hematologic Neoplasms/drug therapy , Puromycin/pharmacology , Aminopeptidases/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , HL-60 Cells , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Humans , Molecular Structure , Puromycin/chemical synthesis , Puromycin/chemistry , Structure-Activity Relationship , Vero Cells
15.
Chem Res Toxicol ; 30(3): 777-784, 2017 03 20.
Article in English | MEDLINE | ID: mdl-28165728

ABSTRACT

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.


Subject(s)
Acetaminophen/toxicity , Chemical and Drug Induced Liver Injury/prevention & control , Disease Models, Animal , Glutathione/pharmacology , Animals , Mice
16.
Invest Ophthalmol Vis Sci ; 57(7): 3231-8, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27333181

ABSTRACT

PURPOSE: To describe a spectral imaging system for small animal studies based on noninvasive endoscopy of the retina, and to present time-resolved spectral changes from live Alzheimer's mice prior to cognitive decline, corroborating our previous in vitro findings. METHODS: Topical endoscope fundus imaging was modified to use a machine vision camera and tunable wavelength system for acquiring monochromatic images across the visible to near-infrared spectral range. Alzheimer's APP/PS1 mice and age-matched, wild-type mice were imaged monthly from months 3 through 8 to assess changes in the fundus reflection spectrum. Optical changes were fit to Rayleigh light scatter models as measures of amyloid aggregation. RESULTS: Good quality spectral images of the central retina were obtained. Short-wavelength reflectance from Alzheimer's mice retinae showed significant reduction over time compared to wild-type mice. Optical changes were consistent with an increase in Rayleigh light scattering in neural retina due to soluble Aß1-42 aggregates. The changes in light scatter showed a monotonic increase in soluble amyloid aggregates over a 6-month period, with significant build up occurring at 7 months. CONCLUSIONS: Hyperspectral imaging technique can be brought inexpensively to the study of retinal changes caused by Alzheimer's disease progression in live small animals. A similar previous finding of reduction in the light reflection over a range of wavelengths in isolated Alzheimer's mice retinae, was reproducible in the living Alzheimer's mice. The technique presented here has a potential for development as an early Alzheimer's retinal diagnostic test in humans, which will support the treatment outcome.


Subject(s)
Alzheimer Disease/complications , Amyloidosis/diagnosis , Early Diagnosis , Endoscopy/methods , Retina/diagnostic imaging , Retinal Diseases/diagnosis , Alzheimer Disease/diagnosis , Amyloidosis/complications , Animals , Diagnosis, Differential , Disease Models, Animal , Follow-Up Studies , Fundus Oculi , Mice , Mice, Transgenic , Reproducibility of Results , Retinal Diseases/etiology
17.
Ann N Y Acad Sci ; 1374(1): 202-9, 2016 06.
Article in English | MEDLINE | ID: mdl-27308865

ABSTRACT

Cyanide is a metabolic poison that inhibits the utilization of oxygen to form ATP. The consequences of acute cyanide exposure are severe; exposure results in loss of consciousness, cardiac and respiratory failure, hypoxic brain injury, and dose-dependent death within minutes to hours. In a mass-casualty scenario, such as an industrial accident or terrorist attack, currently available cyanide antidotes would leave many victims untreated in the short time available for successful administration of a medical countermeasure. This restricted therapeutic window reflects the rate-limiting step of intravenous administration, which requires both time and trained medical personnel. Therefore, there is a need for rapidly acting antidotes that can be quickly administered to large numbers of people. To meet this need, our laboratory is developing sulfanegen, a potential antidote for cyanide poisoning with a novel mechanism based on 3-mercaptopyruvate sulfurtransferase (3-MST) for the detoxification of cyanide. Additionally, sulfanegen can be rapidly administered by intramuscular injection and has shown efficacy in many species of animal models. This article summarizes the journey from concept to clinical leads for this promising cyanide antidote.


Subject(s)
Cyanides/toxicity , Cysteine/analogs & derivatives , Mass Casualty Incidents , Animals , Cysteine/chemistry , Cysteine/pharmacology , Humans , Kinetics , Prodrugs/chemistry , Prodrugs/pharmacology
18.
Redox Biol ; 8: 348-53, 2016 08.
Article in English | MEDLINE | ID: mdl-26972115

ABSTRACT

Glyoxalases prevent the formation of advanced glycation end products by converting glycolysis-derived methylglyoxal to d-lactate with the help of glutathione. Vander Jagt and colleagues previously showed that erythrocytes release about thirty times more d-lactate after infection with the human malaria parasite Plasmodium falciparum. Functional glyoxalases in the host-parasite unit might therefore be crucial for parasite survival. Here, we determined the antimalarial and hemolytic activity of two tight-binding glyoxalase inhibitors using infected and uninfected erythrocytes. In addition, we synthesized and analyzed a set of diester derivates of both tight-binding inhibitors resulting in up to threefold lower IC50 values and an altered methemoglobin formation and hemolytic activity depending on the type of ester. Inhibitor treatments of uninfected erythrocytes revealed an extremely slow inactivation of the host cell glyoxalase, irrespective of inhibitor modifications, and a potential dispensability of the host cell enzyme for parasite survival. Our study highlights the benefits and drawbacks of different esterifications of glutathione-derived inhibitors and demonstrates the suitability of glyoxalase inhibitors as a tool for deciphering the relevance and mode of action of different glyoxalase systems in a host-parasite unit.


Subject(s)
Erythrocytes/drug effects , Host-Parasite Interactions/drug effects , Lactoylglutathione Lyase/antagonists & inhibitors , Malaria, Falciparum/drug therapy , Animals , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemical synthesis , Erythrocytes/parasitology , Glutathione/metabolism , Glycation End Products, Advanced/drug effects , Glycation End Products, Advanced/metabolism , Hemolysis/drug effects , Humans , Lactoylglutathione Lyase/genetics , Malaria, Falciparum/genetics , Malaria, Falciparum/parasitology , Plasmodium falciparum/drug effects , Plasmodium falciparum/pathogenicity
20.
ACS Chem Neurosci ; 6(2): 306-15, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25354367

ABSTRACT

Amyloidopathic disorders such as Alzheimer's disease present symptomology years after the entrenchment of amyloidogenic imbalance. The pathologic α-helix → ß-strand conversion of amyloid ß(1-42) and amyloid ß(1-40) peptides causes neuronal death in the vicinity. Symptomology often presents only after significant neurodegeneration. This thus warrants early detection of amyloidopathy in Alzheimer's disease. Nonexistent modalities for direct identification and quantitation of soluble amyloid aggregates or (proto)fibrils forced us to undertake the development of a spectrophotometric technique to support ongoing drug design. Key requirements were independence from the need for extraneous staining, unambiguous amyloid aggregate detection, and minimal influence of interpretative errors. A Cytoviva instrument pivotal to this study captures scattering of light of visible-near-infrared (VNIR, 400-1000 nm) wavelengths within each pixel of the microscopic view field. We thus assembled a scattering intensity pattern database that provided "signatures" of amyloid aggregates. Comparison of unknown samples against this database enabled direct detection of amyloid aggregates. The technique was found useful for monitoring retinal and brain amyloidopathy in an ongoing preclinical anti-AD study, attesting to the technique's sensitivity and specificity. Interestingly, the technique was found applicable not just to excised brain tissue but also to isolated mouse retina. With the retina being heralded widely as a (diagnostic) extension of the CNS and retinal amyloidopathy occurring well before that in the brain, this development raises a possibility for the first direct retinal imaging diagnosis of early asymptomatic Alzheimer's disease.


Subject(s)
Alzheimer Disease/diagnosis , Alzheimer Disease/metabolism , Amyloidosis/metabolism , Retina/metabolism , Spectrophotometry/methods , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloidosis/pathology , Animals , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Cognition Disorders/metabolism , Cognition Disorders/pathology , Cytoplasm/metabolism , Disease Models, Animal , Female , Fluorescent Antibody Technique , Humans , Mice, Inbred C57BL , Mice, Transgenic , Models, Neurological , Nootropic Agents/pharmacology , Peptide Fragments/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism , Retina/pathology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...