Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Nitric Oxide ; 82: 59-74, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30394348

ABSTRACT

The development of small molecule modulators of NO/cGMP signaling for use in the CNS has lagged far behind the use of such clinical agents in the periphery, despite the central role played by NO/cGMP in learning and memory, and the substantial evidence that this signaling pathway is perturbed in neurodegenerative disorders, including Alzheimer's disease. The NO-chimeras, NMZ and Nitrosynapsin, have yielded beneficial and disease-modifying responses in multiple preclinical animal models, acting on GABAA and NMDA receptors, respectively, providing additional mechanisms of action relevant to synaptic and neuronal dysfunction. Several inhibitors of cGMP-specific phosphodiesterases (PDE) have replicated some of the actions of these NO-chimeras in the CNS. There is no evidence that nitrate tolerance is a phenomenon relevant to the CNS actions of NO-chimeras, and studies on nitroglycerin in the periphery continue to challenge the dogma of nitrate tolerance mechanisms. Hybrid nitrates have shown much promise in the periphery and CNS, but to date only one treatment has received FDA approval, for glaucoma. The potential for allosteric modulation of soluble guanylate cyclase (sGC) in brain disorders has not yet been fully explored nor exploited; whereas multiple applications of PDE inhibitors have been explored and many have stalled in clinical trials.


Subject(s)
Central Nervous System Agents/pharmacology , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/metabolism , Cyclic GMP/metabolism , Drug Discovery , Nitric Oxide/metabolism , Small Molecule Libraries/pharmacology , Animals , Central Nervous System Agents/chemical synthesis , Central Nervous System Agents/chemistry , Humans , Signal Transduction/drug effects , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry
2.
Cephalalgia ; 38(8): 1471-1484, 2018 07.
Article in English | MEDLINE | ID: mdl-29022756

ABSTRACT

Background Nitric oxide (NO) has been heavily implicated in migraine. Nitroglycerin is a prototypic NO-donor, and triggers migraine in humans. However, nitroglycerin also induces oxidative/nitrosative stress and is a source of peroxynitrite - factors previously linked with migraine etiology. Soluble guanylyl cyclase (sGC) is the high affinity NO receptor in the body, and the aim of this study was to identify the precise role of sGC in acute and chronic migraine. Methods We developed a novel brain-bioavailable sGC stimulator (VL-102), and tested its hyperalgesic properties in mice. We also determined the effect of VL-102 on c-fos and calcitonin gene related peptide (CGRP) immunoreactivity within the trigeminovascular complex. In addition, we also tested the known sGC inhibitor, ODQ, within the chronic nitroglycerin migraine model. Results VL-102-evoked acute and chronic mechanical cephalic and hind-paw allodynia in a dose-dependent manner, which was blocked by the migraine medications sumatriptan, propranolol, and topiramate. In addition, VL-102 also increased c-fos and CGRP expressing cells within the trigeminovascular complex. Importantly, ODQ completely inhibited acute and chronic hyperalgesia induced by nitroglycerin. ODQ also blocked hyperalgesia already established by chronic nitroglycerin, implicating this pathway in migraine chronicity. Conclusions These results indicate that nitroglycerin causes migraine-related pain through stimulation of the sGC pathway, and that super-activation of this receptor may be an important component for the maintenance of chronic migraine. This work opens the possibility for negative sGC modulators as novel migraine therapies.


Subject(s)
Hyperalgesia/chemically induced , Hyperalgesia/enzymology , Migraine Disorders/enzymology , Soluble Guanylyl Cyclase/physiology , Adrenergic beta-Antagonists/administration & dosage , Adrenergic beta-Antagonists/therapeutic use , Allosteric Regulation , Animals , Anticonvulsants/administration & dosage , Anticonvulsants/therapeutic use , Calcitonin Gene-Related Peptide/biosynthesis , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/therapeutic use , Female , Hyperalgesia/drug therapy , Male , Mice , Mice, Inbred C57BL , Migraine Disorders/chemically induced , Migraine Disorders/drug therapy , Migraine Disorders/etiology , Molecular Targeted Therapy , Nitric Oxide/adverse effects , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Nitroglycerin/pharmacology , Oxadiazoles/administration & dosage , Oxadiazoles/metabolism , Oxadiazoles/therapeutic use , Propranolol/administration & dosage , Propranolol/therapeutic use , Proto-Oncogene Proteins c-fos/biosynthesis , Quinoxalines/administration & dosage , Quinoxalines/metabolism , Quinoxalines/therapeutic use , Serotonin 5-HT1 Receptor Agonists/administration & dosage , Serotonin 5-HT1 Receptor Agonists/therapeutic use , Soluble Guanylyl Cyclase/metabolism , Sumatriptan/administration & dosage , Sumatriptan/therapeutic use , Topiramate/administration & dosage , Topiramate/therapeutic use
3.
J Biol Chem ; 289(44): 30538-30555, 2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25217640

ABSTRACT

Previous data demonstrate that bexarotene (Bex), retinoid X receptor (RXR) agonist, reduces soluble and insoluble amyloid-ß (Aß) in Alzheimer disease (AD)-transgenic mice either by increasing the levels of mouse apolipoprotein E (apoE) or increasing ABCA1/ABCG1-induced apoE lipoprotein association/lipidation. However, although the mechanism of action of RXR agonists remains unclear, a major concern for their use is human (h)-APOE4, the greatest AD genetic risk factor. If APOE4 imparts a toxic gain-of-function, then increasing apoE4 may increase soluble Aß, likely the proximal AD neurotoxin. If the APOE4 loss-of-function is lipidation of apoE4, then induction of ABCA1/ABCG1 may be beneficial. In novel EFAD-Tg mice (overexpressing h-Aß42 with h-APOE), levels of soluble Aß (Aß42 and oligomeric Aß) are highest in E4FAD hippocampus (HP) > E3FAD-HP > E4FAD cortex (CX) > E3FAD-CX, whereas levels of lipoprotein-associated/lipidated apoE have the opposite pattern (6 months). In E4FAD-HP, short-term RXR agonist treatment (Bex or LG100268; 5.75-6 months) increased ABCA1, apoE4 lipoprotein-association/lipidation, and apoE4/Aß complex, decreased soluble Aß, and increased PSD95. In addition, hydrogel delivery, which mimics low sustained release, was equally effective as gavage for Bex and LG100268. RXR agonists induced no beneficial effects in the E4FAD-HP in a prevention protocol (5-6 months) and actually increased soluble Aß levels in E3FAD-CX and E4FAD-CX with the short-term protocol, possibly the result of systemic hepatomegaly. Thus, RXR agonists address the loss-of-function associated with APOE4 and exacerbated by Aß pathology, i.e. low levels of apoE4 lipoprotein association/lipidation. Further studies are vital to address whether RXR agonists are an APOE4-specific AD therapeutic and the systemic side effects that limit translational application.


Subject(s)
Amyloid beta-Peptides/metabolism , Apolipoproteins E/genetics , Nicotinic Acids/administration & dosage , Peptide Fragments/metabolism , Retinoid X Receptors/agonists , Tetrahydronaphthalenes/administration & dosage , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/metabolism , Administration, Oral , Alzheimer Disease/drug therapy , Animals , Bexarotene , Disks Large Homolog 4 Protein , Drug Evaluation, Preclinical , Genotype , Guanylate Kinases/metabolism , Humans , Lipoproteins/metabolism , Liver/drug effects , Liver/pathology , Male , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Nicotinic Acids/adverse effects , Nicotinic Acids/pharmacokinetics , Organ Size/drug effects , Retinoid X Receptors/metabolism , Solubility , Tetrahydronaphthalenes/adverse effects , Tetrahydronaphthalenes/pharmacokinetics
4.
BMC Neurosci ; 16: 67, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26480871

ABSTRACT

BACKGROUND: Synaptic dysfunction is a key event in pathogenesis of neurodegenerative diseases such as Alzheimer's disease (AD) where synapse loss pathologically correlates with cognitive decline and dementia. Although evidence suggests that aberrant protein production and aggregation are the causative factors in familial subsets of such diseases, drugs singularly targeting these hallmark proteins, such as amyloid-ß, have failed in late stage clinical trials. Therefore, to provide a successful disease-modifying compound and address synaptic dysfunction and memory loss in AD and mixed pathology dementia, we repurposed a clinically proven drug, CMZ, with neuroprotective and anti-inflammatory properties via addition of nitric oxide (NO) and cGMP signaling property. RESULTS: The novel compound, NMZ, was shown to retain the GABAA potentiating actions of CMZ in vitro and sedative activity in vivo. Importantly, NMZ restored LTP in hippocampal slices from AD transgenic mice, whereas CMZ was without effect. NMZ reversed amnestic blockade of acetylcholine receptors by scopolamine as well as NMDA receptor blockade by a benzodiazepine and a NO synthase inhibitor in the step-through passive avoidance (STPA) test of learning and working memory. A PK/PD relationship was developed based on STPA analysis coupled with pharmacokinetic measures of drug levels in the brain: at 1 nM concentration in brain and plasma, NMZ was able to restore memory consolidation in mice. CONCLUSION: Our findings show that NMZ embodies a promising pharmacological approach targeting synaptic dysfunction and opens new avenues for neuroprotective intervention strategies in mixed pathology AD, neurodegeneration, and dementia.


Subject(s)
Alzheimer Disease/drug therapy , Chlormethiazole/analogs & derivatives , Drug Repositioning/methods , GABA-A Receptor Agonists/pharmacology , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Neuroprotective Agents/pharmacology , Nootropic Agents/pharmacology , Animals , CREB-Binding Protein/metabolism , Cyclic GMP/metabolism , Disease Models, Animal , GABA-A Receptor Agonists/pharmacokinetics , Male , Mice , Mice, Transgenic , Neuroprotective Agents/pharmacokinetics , Nitric Oxide/metabolism , Nootropic Agents/pharmacokinetics , Signal Transduction/drug effects , Synapses/drug effects , Synapses/pathology , Xenopus laevis
5.
Antioxidants (Basel) ; 11(2)2022 Jan 24.
Article in English | MEDLINE | ID: mdl-35204103

ABSTRACT

To evaluate the differences in action of commercially available 2-oxoglutarate mimetics and "branched-tail" oxyquinoline inhibitors of hypoxia-inducible factor prolyl hydroxylase (HIF PHD), the inhibitors' IC50 values in the activation of HIF1 ODD-luciferase reporter were selected for comparative transcriptomics. Structure-activity relationship and computer modeling for the oxyquinoline series of inhibitors led to the identification of novel inhibitors, which were an order of magnitude more active in the reporter assay than roxadustat and vadadustat. Unexpectedly, 2-methyl-substitution in the oxyquinoline core of the best HIF PHD inhibitor was found to be active in the reporter assay and almost equally effective in the pretreatment paradigm of the oxygen-glucose deprivation in vitro model. Comparative transcriptomic analysis of the signaling pathways induced by HIF PHD inhibitors showed high potency of the two novel oxyquinoline inhibitors (#4896-3249 and #5704-0720) at 2 µM concentrations matching the effect of 30 µM roxadustat and 500 µM dimethyl oxalyl glycine in inducing HIF1 and HIF2-linked pathways. The two oxyquinoline inhibitors exerted the same activation of HIF-triggered glycolytic pathways but opposite effects on signaling pathways linked to alternative substrates of HIF PHD 1 and 3, such as p53, NF-κB, and ATF4. This finding can be interpreted as the specificity of the 2-methyl-substitute variant for HIF PHD2.

6.
ACS Pharmacol Transl Sci ; 4(1): 143-154, 2021 Feb 12.
Article in English | MEDLINE | ID: mdl-33615168

ABSTRACT

Selective liver X receptor (LXR) agonists have been extensively pursued as therapeutics for Alzheimer's disease and related dementia (ADRD) and, for comorbidities such as type 2 diabetes (T2D) and cerebrovascular disease (CVD), disorders with underlying impaired insulin signaling, glucose metabolism, and cholesterol mobilization. The failure of the LXR-focused approach led us to pursue a novel strategy to discover nonlipogenic ATP-binding cassette transporter A1 (ABCA1) inducers (NLAIs): screening for ABCA1-luciferase activation in astrocytoma cells and counterscreening against lipogenic gene upregulation in hepatocarcinoma cells. Beneficial effects of LXRß agonists mediated by ABCA1 include the following: control of cholesterol and phospholipid efflux to lipid-poor apolipoproteins forming beneficial peripheral HDL and HDL-like particles in the brain and attenuation of inflammation. While rare, ABCA1 variants reduce plasma HDL and correlate with an increased risk of ADRD and CVD. In secondary assays, NLAI hits enhanced cholesterol mobilization and positively impacted in vitro biomarkers associated with insulin signaling, inflammatory response, and biogenic properties. In vivo target engagement was demonstrated after oral administration of NLAIs in (i) mice fed a high-fat diet, a model for obesity-linked T2D, (ii) mice administered LPS, and (iii) mice with accelerated oxidative stress. The lack of adverse effects on lipogenesis and positive effects on multiple biomarkers associated with T2D and ADRD supports this novel phenotypic approach to NLAIs as a platform for T2D and ADRD drug discovery.

7.
Redox Biol ; 32: 101486, 2020 05.
Article in English | MEDLINE | ID: mdl-32155582

ABSTRACT

Oxidative stress induced by lipid peroxidation products (LPP) accompanies aging and has been hypothesized to exacerbate the secondary cascade in traumatic brain injury (TBI). Increased oxidative stress is a contributor to loss of neural reserve that defines the ability to maintain healthy cognitive function despite the accumulation of neuropathology. ALDH2-/- mice are unable to clear aldehyde LPP by mitochondrial aldehyde dehydrogenase-2 (Aldh2) detoxification and provide a model to study mild TBI (mTBI), therapeutic interventions, and underlying mechanisms. The ALDH2-/- mouse model presents with elevated LPP-mediated protein modification, lowered levels of PSD-95, PGC1-α, and SOD-1, and mild cognitive deficits from 4 months of age. LPP scavengers are neuroprotective in vitro and in ALDH2-/- mice restore cognitive performance. A single-hit, closed skull mTBI failed to elicit significant effects in WT mice; however, ALDH2-/- mice showed a significant inflammatory cytokine surge in the ipsilateral hemisphere 24 h post-mTBI, and increased GFAP cleavage, a biomarker for TBI. Known neuroprotective agents, were able to reverse the effects of mTBI. This new preclinical model of mTBI, incorporating significant perturbations in behavior, inflammation, and clinically relevant biomarkers, allows mechanistic study of the interaction of LPP and neurotrauma in loss of neural reserve.


Subject(s)
Brain Concussion , Neuroprotective Agents , Aldehyde Dehydrogenase, Mitochondrial/genetics , Animals , Disease Models, Animal , Mice , Oxidative Stress
8.
ACS Chem Neurosci ; 9(5): 894-900, 2018 05 16.
Article in English | MEDLINE | ID: mdl-29338172

ABSTRACT

Activation of HIF-1α and Nrf2 is a primary component of cellular response to oxidative stress, and activation of HIF-1α and Nrf2 provides neuroprotection in models of neurodegenerative disorders, including ischemic stroke, Alzheimer's and Parkinson's diseases. Screening a library of CNS-targeted drugs using novel reporters for HIF-1α and Nrf2 elevation in neuronal cells revealed histone deacetylase (HDAC) inhibitors as potential activators of these pathways. We report the identification of phenylhydroxamates as single agents exhibiting tripartite inhibition of HDAC6, inhibition of HIF-1 prolyl hydroxylase (PHD), and activation of Nrf2. Two superior tripartite agents, ING-6 and ING-66, showed neuroprotection against various cellular insults, associated with stabilization of both Nrf2 and HIF-1, and expression of their respective target genes in vitro and in vivo. Discovery of the innate ability of phenylhydroxamate HDAC inhibitors to activate Nrf2 and HIF provides a novel route to multifunctional neuroprotective agents and cautions against HDAC6 selective inhibitors as chemical probes of specific HDAC isoform function.


Subject(s)
Histone Deacetylase 6/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxylamines/pharmacology , Neurodegenerative Diseases/drug therapy , Neurons/drug effects , Neuroprotective Agents/pharmacology
9.
Curr Med Chem ; 23(24): 2770-2788, 2016.
Article in English | MEDLINE | ID: mdl-27776476

ABSTRACT

cAMP-response element-binding protein (CREB) plays a central role in various aspects of central nervous system (CNS) function, ranging from the developmental stages to neuronal plasticity and survival in adult brain. Activation of CREB plays a crucial role in learning and memory and is at the convergence of multiple intracellular signaling cascades including CAMKII and MAPK. This review focuses on the important functions of nitric oxide (NO) in activating CREB via the NO receptor, soluble guanylyl cyclase (sGC), and production of the second messenger, cGMP. The involvement of the NO/cGMP signaling pathway in synaptic plasticity suggests several avenues for therapeutic intervention, and targeting early synaptic degeneration could be an attractive approach for the development of novel disease-modifying approaches to treat cognition and memory dysfunction in neurodegenerative diseases.


Subject(s)
Alzheimer Disease/pathology , Central Nervous System/metabolism , Cyclic GMP/metabolism , Neurodegenerative Diseases/pathology , Nitric Oxide/metabolism , Alzheimer Disease/metabolism , CREB-Binding Protein/metabolism , Humans , Neurodegenerative Diseases/metabolism , Neuronal Plasticity/physiology , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/metabolism , Signal Transduction , Soluble Guanylyl Cyclase/antagonists & inhibitors , Soluble Guanylyl Cyclase/metabolism , Synapses/metabolism
10.
Mol Neurodegener ; 11: 35, 2016 Apr 29.
Article in English | MEDLINE | ID: mdl-27129593

ABSTRACT

BACKGROUND: Clinical failures singularly targeting amyloid-ß pathology indicate a critical need for alternative Alzheimer's disease (AD) therapeutic strategies. The mixed pathology reported in a large population of AD patients demands a multifunctional drug approach. Since activation of cAMP response element binding protein (CREB) plays a crucial role in synaptic strengthening and memory formation, we retooled a clinical drug with known neuroprotective and anti-inflammatory activity to activate CREB, and validated this novel multifunctional drug, NMZ, in 4 different mouse models of AD. RESULTS: NMZ was tested in three mouse models of familial AD and one model of sporadic AD. In 3 × Tg hippocampal slices, NMZ restored LTP. In vivo, memory was improved with NMZ in all animal models with robust cognitive deficits. NMZ treatment lowered neurotoxic forms of Aß in both APP/PS1 and 3 × Tg transgenic mice while also restoring neuronal plasticity biomarkers in the 3 × Tg mice. In EFAD mice, incorporation of the major genetic AD risk factor, hAPOE4, did not mute the beneficial drug effects. In a novel sporadic mouse model that manifests AD-like pathology caused by accelerated oxidative stress in the absence of any familial AD mutation, oral administration of NMZ attenuated hallmark AD pathology and restored biomarkers of synaptic and neuronal function. CONCLUSIONS: The multifunctional approach, embodied by NMZ, was successful in mouse models of AD incorporating Aß pathology (APP/PS1), tau pathology (3xTg), and APOE4, the major human genetic risk factor for AD (EFAD). The efficacy observed in a novel model of sporadic AD (Aldh2 (-/-) ) demonstrates that the therapeutic approach is not limited to rare, familial AD genetic mutations. The multifunctional drug, NMZ, was not designed directly to target Aß and tau pathology; however, the attenuation of this hallmark pathology suggests the approach to be a highly promising, disease-modifying strategy for AD and mixed pathology dementia.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Protein Precursor/metabolism , Cognition Disorders/metabolism , Memory/physiology , Neuronal Plasticity/physiology , tau Proteins/metabolism , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Hippocampus/metabolism , Mice, Knockout , Mice, Transgenic
11.
Biomaterials ; 35(36): 9635-48, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25154665

ABSTRACT

Peripheral artery disease (PAD) is characterized by vessel occlusion and ischemia in the limbs. Treatment for PAD with surgical interventions has been showing limited success. Moreover, recent clinical trials with treatment of angiogenic growth factors proved ineffective as increased angiogenesis triggered severe inflammation in a proportionally coupled fashion. Hence, the overarching goal of this research was to address this issue by developing a biomaterial system that enables controlled, dual delivery of pro-angiogenic C16 and anti-inflammatory Ac-SDKP peptides in a minimally-invasive way. To achieve the goal, a peptide-loaded injectable microgel system was developed and tested in a mouse model of PAD. When delivered through multiple, low volume injections, the combination of C16 and Ac-SDKP peptides promoted angiogenesis, muscle regeneration, and perfusion recovery, while minimizing detrimental inflammation. Additionally, this peptide combination regulated inflammatory TNF-α pathways independently of MMP-9 mediated pathways of angiogenesis in vitro, suggesting a potential mechanism by which angiogenic and inflammatory responses can be uncoupled in the context of PAD. This study demonstrates a translatable potential of the dual peptide-loaded injectable microgel system for PAD treatment.


Subject(s)
Angiogenesis Inducing Agents/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Drug Carriers/chemistry , Inflammation/drug therapy , Neovascularization, Physiologic/drug effects , Oligopeptides/administration & dosage , Peripheral Arterial Disease/drug therapy , Angiogenesis Inducing Agents/therapeutic use , Animals , Anti-Inflammatory Agents/therapeutic use , Cell Line , Human Umbilical Vein Endothelial Cells , Inflammation/complications , Inflammation/immunology , Injections , Matrix Metalloproteinase 9/immunology , Matrix Metalloproteinase Inhibitors/administration & dosage , Matrix Metalloproteinase Inhibitors/therapeutic use , Mice , Oligopeptides/therapeutic use , Peripheral Arterial Disease/complications , Peripheral Arterial Disease/immunology , Polyesters/chemistry , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology
SELECTION OF CITATIONS
SEARCH DETAIL