Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Bioorg Med Chem Lett ; 92: 129409, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37453616

ABSTRACT

Phosphodiesterase 5 (PDE5) is a cyclic guanosine monophosphate-degrading enzyme involved in numerous biological pathways. Inhibitors of PDE5 are important therapeutics for the treatment of neurodegenerative diseases, including Alzheimer's disease (AD). We previously reported the first generation of quinoline-based PDE5 inhibitors for the treatment of AD. However, the short in vitro microsomal stability rendered them unsuitable drug candidates. Here we report a series of new quinoline-based PDE5 inhibitors. Among them, compound 4b, 8-cyclopropyl-3-(hydroxymethyl)-4-(((6-methoxypyridin-3-yl)methyl)amino)quinoline-6-carbonitrile, shows a PDE5 IC50 of 20 nM and improved in vitro microsomal stability (t1/2 = 44.6 min) as well as excellent efficacy in restoring long-term potentiation, a type of synaptic plasticity to underlie memory formation, in electrophysiology experiments with a mouse model of AD. These results provide an insight into the development of a new class of PDE5 inhibitors for the treatment of AD.


Subject(s)
Alzheimer Disease , Quinolines , Mice , Animals , Phosphodiesterase 5 Inhibitors/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Neuronal Plasticity , Alzheimer Disease/drug therapy , Quinolines/pharmacology , Quinolines/therapeutic use
2.
Biochem Pharmacol ; 176: 113818, 2020 06.
Article in English | MEDLINE | ID: mdl-31978378

ABSTRACT

Nitric oxide (NO) is a gaseous molecule that plays a multifactorial role in several cellular processes. In the central nervous system, the NO dual nature in neuroprotection and neurotoxicity has been explored to unveil its involvement in Alzheimer's disease (AD). A growing body of research shows that the activation of the NO signaling pathway leading to the phosphorylation of the transcription factor cyclic adenine monophosphate responsive element binding protein (CREB) (so-called NO/cGMP/PKG/CREB signaling pathway) ameliorates altered neuroplasticity and memory deficits in AD animal models. In addition to NO donors, several other pharmacological agents, such as phosphodiesterase 5 (PDE5) inhibitors have been used to activate the pathway and rescue memory disorders. PDE5 inhibitors, including sildenafil, tadalafil and vardenafil, are marketed for the treatment of erectile dysfunction and arterial pulmonary hypertension due to their vasodilatory properties. The ability of PDE5 inhibitors to interfere with the NO/cGMP/PKG/CREB signaling pathway by increasing the levels of cGMP has prompted the hypothesis that PDE5 inhibition might be used as an effective therapeutic strategy for the treatment of AD. To this end, newly designed PDE5 inhibitors belonging to different chemical classes with improved pharmacologic profile (e.g. higher potency, improved selectivity, and blood-brain barrier penetration) have been synthesized and evaluated in several animal models of AD. In addition, recent medicinal chemistry effort has led to the development of agents concurrently acting on the PDE5 enzyme and a second target involved in AD. Both marketed and investigational PDE5 inhibitors have shown to reverse cognitive defects in young and aged wild type mice as well as transgenic mouse models of AD and tauopathy using a variety of behavioral tasks. These studies confirmed the therapeutic potential of PDE5 inhibitors as cognitive enhancers. However, clinical studies assessing cognitive functions using marketed PDE5 inhibitors have not been conclusive. Drug discovery efforts by our group and others are currently directed towards the development of novel PDE5 inhibitors tailored to AD with improved pharmacodynamic and pharmacokinetic properties. In summary, the present perspective reports an overview of the correlation between the NO signaling and AD, as well as an outline of the PDE5 inhibitors used as an alternative approach in altering the NO pathway leading to an improvement of learning and memory. The last two sections describe the preclinical and clinical evaluation of PDE5 inhibitors for the treatment of AD, providing a comprehensive analysis of the current status of the AD drug discovery efforts involving PDE5 as a new therapeutic target.


Subject(s)
Alzheimer Disease/drug therapy , Disease Models, Animal , Phosphodiesterase 5 Inhibitors/therapeutic use , Signal Transduction/drug effects , Alzheimer Disease/enzymology , Alzheimer Disease/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Humans , Nitric Oxide/metabolism
3.
Mol Neurodegener ; 14(1): 26, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31248451

ABSTRACT

BACKGROUND: Soluble aggregates of oligomeric forms of tau protein (oTau) have been associated with impairment of synaptic plasticity and memory in Alzheimer's disease. However, the molecular mechanisms underlying the synaptic and memory dysfunction induced by elevation of oTau are still unknown. METHODS: This work used a combination of biochemical, electrophysiological and behavioral techniques. Biochemical methods included analysis of phosphorylation of the cAMP-responsive element binding (CREB) protein, a transcriptional factor involved in memory, histone acetylation, and expression immediate early genes c-Fos and Arc. Electrophysiological methods included assessment of long-term potentiation (LTP), a type of synaptic plasticity thought to underlie memory formation. Behavioral studies investigated both short-term spatial memory and associative memory. These phenomena were examined following oTau elevation. RESULTS: Levels of phospho-CREB, histone 3 acetylation at lysine 27, and immediate early genes c-Fos and Arc, were found to be reduced after oTau elevation during memory formation. These findings led us to explore whether up-regulation of various components of the nitric oxide (NO) signaling pathway impinging onto CREB is capable of rescuing oTau-induced impairment of plasticity, memory, and CREB phosphorylation. The increase of NO levels protected against oTau-induced impairment of LTP through activation of soluble guanylyl cyclase. Similarly, the elevation of cGMP levels and stimulation of the cGMP-dependent protein kinases (PKG) re-established normal LTP after exposure to oTau. Pharmacological inhibition of cGMP degradation through inhibition of phosphodiesterase 5 (PDE5), rescued oTau-induced LTP reduction. These findings could be extrapolated to memory because PKG activation and PDE5 inhibition rescued oTau-induced memory impairment. Finally, PDE5 inhibition re-established normal elevation of CREB phosphorylation and cGMP levels after memory induction in the presence of oTau. CONCLUSIONS: Up-regulation of CREB activation through agents acting on the NO cascade might be beneficial against tau-induced synaptic and memory dysfunctions.


Subject(s)
Alzheimer Disease/metabolism , Long-Term Potentiation/physiology , Neuronal Plasticity/physiology , Nitric Oxide/metabolism , tau Proteins/metabolism , Amyloid beta-Peptides/metabolism , Animals , Female , Male , Memory/physiology , Memory Disorders/metabolism , Mice, Inbred C57BL , Neurons/metabolism
5.
J Med Chem ; 60(21): 8858-8875, 2017 11 09.
Article in English | MEDLINE | ID: mdl-28985058

ABSTRACT

Phosphodiesterase 5 (PDE5) hydrolyzes cyclic guanosine monophosphate (cGMP) leading to increased levels of the cAMP response element binding protein (CREB), a transcriptional factor involved with learning and memory processes. We previously reported potent quinoline-based PDE5 inhibitors (PDE5Is) for the treatment of Alzheimer's disease (AD). However, the low aqueous solubility rendered them undesirable drug candidates. Here we report a series of novel PDE5Is with two new scaffolds, 1,2,3,4-tetrahydrobenzo[b][1,6]naphthyridine and 2,3-dihydro-1H-pyrrolo[3,4-b]quinolin-1-one. Among them, compound 6c, 2-acetyl-10-((3-chloro-4-methoxybenzyl)amino)-1,2,3,4-tetrahydrobenzo[b][1,6]naphthyridine-8-carbonitrile, the most potent compound, has an excellent in vitro IC50 (0.056 nM) and improved aqueous solubility as well as good efficacy in a mouse model of AD. Furthermore, we are proposing two plausible binding modes obtained through in silico docking, which provide insights into the structural basis of the activity of the two series of compounds reported herein.


Subject(s)
Alzheimer Disease/drug therapy , Naphthyridines/chemical synthesis , Phosphodiesterase 5 Inhibitors/chemical synthesis , Animals , Binding Sites , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic GMP/metabolism , Humans , Mice , Molecular Docking Simulation , Naphthyridines/therapeutic use , Phosphodiesterase 5 Inhibitors/therapeutic use , Quinolines , Solubility , Structure-Activity Relationship
6.
Cent Nerv Syst Agents Med Chem ; 17(2): 129-134, 2017.
Article in English | MEDLINE | ID: mdl-27554921

ABSTRACT

BACKGROUND: MRJF4, a novel haloperidol metabolite II prodrug, was obtained through the esterification of the secondary hydroxyl group of haloperidol metabolite II with 4-phenylbutyric acid. The activities of (±)-MRJF4 and its two enantiomers [(+)-MRJF4 and (-)-MRJF4] as tumor specific inducers of pro-apoptotic genes were evaluated on malignant C6 glioma cells. In particular, changes in Nf-κB signaling pathway, activity of nitric oxide synthases (NOS), metalloproteinases (MMPs), and membrane adhesion proteins were investigated. RESULTS: IκBα reduced phosphorylation and iNOS lowered activity could be correlated with the previously demonstrated decreased proliferation and tumor progression of C6 cells upon 24 h of treatment with all the three compounds. Integrin ß1 decreased expression, at the same experimental time, seems to support lower C6 cells migrative capability and the consequent reduced invasiveness of these cells upon treatment with (±)-MRJF4 and its enantiomers. CONCLUSION: These results suggest that this multi-target prodrug and its two enantiomers might be a valuable clinical tool for the treatment of metastatic glioblastoma.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Glioma/metabolism , Haloperidol/analogs & derivatives , Phenylbutyrates/pharmacology , Prodrugs/pharmacology , Animals , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Glioma/drug therapy , Glioma/pathology , Haloperidol/pharmacology , Haloperidol/therapeutic use , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Phenylbutyrates/therapeutic use , Prodrugs/therapeutic use , Rats
7.
J Alzheimers Dis ; 52(1): 295-302, 2016.
Article in English | MEDLINE | ID: mdl-26967220

ABSTRACT

Phosphodiesterase 5 (PDE5) is a critical component of the cGMP-PKG axis of cellular signaling in neurons, and inhibition of PDE5 has been shown to be therapeutic in a wide range of neurologic conditions in animal models. However, enthusiasm for PDE5 inhibitors in humans is limited by data suggesting that PDE5 may not exist in human neurons. Here, we first show that past attempts to quantify PDE5 mRNA were flawed due to the use of incorrect primers, and that when correct primers are used, PDE5 mRNA is detectable in human brain tissue. We then show that PDE5 protein exists in human brain by western blot and ELISA. Most importantly, we performed immunohistochemistry and demonstrate that PDE5 is present in human neurons. We hope that this work will trigger a renewed interest in the development of PDE5 inhibitors for neurologic disease.


Subject(s)
Brain/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Neurons/enzymology , Blotting, Western , DNA Primers , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Nervous System Diseases/drug therapy , Phosphodiesterase 5 Inhibitors/therapeutic use , Polymerase Chain Reaction , RNA, Messenger/metabolism
8.
J Med Chem ; 59(21): 9960-9966, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27739690

ABSTRACT

(±)-MRJF22 [(±)-2], a novel prodrug of haloperidol metabolite II (sigma-1 receptor antagonist/sigma-2 receptor agonist ligand) obtained by conjugation to valproic acid (histone deacetylase inhibitor) via an ester bond, exhibits antiangiogenic activity, being able to reduce human retinal endothelial cell (HREC) viability in a comparable manner to bevacizumab. Moreover, (±)-2 was able to significantly reduce viable cells count, endothelial cell migration, and tube formation in vascular endothelial growth factor A (VEGF-A) stimulated HREC cultures.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endothelial Cells/drug effects , Haloperidol/pharmacology , Microvessels/drug effects , Neovascularization, Physiologic/drug effects , Retina/cytology , Valproic Acid/pharmacology , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/chemistry , Cell Survival/drug effects , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Haloperidol/chemical synthesis , Haloperidol/chemistry , Humans , Molecular Structure , Structure-Activity Relationship , Valproic Acid/chemical synthesis , Valproic Acid/chemistry
9.
Neurotherapeutics ; 12(1): 29-41, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25575647

ABSTRACT

Therapeutic attempts to cure Alzheimer's disease (AD) have failed, and new strategies are desperately needed. Motivated by this reality, many laboratories (including our own) have focused on synaptic dysfunction in AD because synaptic changes are highly correlated with the severity of clinical dementia. In particular, memory formation is accompanied by altered synaptic strength, and this phenomenon (and its dysfunction in AD) has been a recent focus for many laboratories. The molecule cyclic adenosine monophosphate response element-binding protein (CREB) is at a central converging point of pathways and mechanisms activated during the processes of synaptic strengthening and memory formation, as CREB phosphorylation leads to transcription of memory-associated genes. Disruption of these mechanisms in AD results in a reduction of CREB activation with accompanying memory impairment. Thus, it is likely that strategies aimed at these mechanisms will lead to future therapies for AD. In this review, we will summarize literature that investigates 5 possible therapeutic pathways for rescuing synaptic dysfunction in AD: 4 enzymatic pathways that lead to CREB phosphorylation (the cyclic adenosine monophosphate cascade, the serine/threonine kinases extracellular regulated kinases 1 and 2, the nitric oxide cascade, and the calpains), as well as histone acetyltransferases and histone deacetylases (2 enzymes that regulate the histone acetylation necessary for gene transcription).


Subject(s)
Alzheimer Disease/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Synaptic Transmission/physiology , Alzheimer Disease/pathology , Animals , Humans , Synapses/metabolism , Synapses/pathology
10.
Eur J Med Chem ; 90: 1-9, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25461306

ABSTRACT

In a previous work we reported the antiproliferative effects of (±)-MRJF4, a novel haloperidol metabolite II (HP-mII) (a sigma-1 antagonist and sigma-2 agonist) prodrug, obtained through conjugation to 4-phenylbutyric acid (PhBA) [a histone deacetylase inhibitor (HDACi)] via an ester bond. As a continuation of this work, here we report the asymmetric synthesis of compounds (R)-(+)-MRJF4 and (S)-(-)-MRJF4 and the evaluation of their biological activity on rat C6 glioma cells, derived from glioblastoma multiforme (GBM), which is the most common and deadliest central nervous system (CNS) invasive malignancy. Favourable physicochemical properties, high permeability in the parallel artificial membrane permeability assay (PAMPA), good enzymatic and chemical stability, in vivo anticancer activity, associated with the capacity to reduce cell viability and to increase cell death by apoptosis, render compound (R)-(+)-MRJF4 a promising candidate for the development of a useful therapeutic for gliomas therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Glioma/drug therapy , Haloperidol/pharmacology , Prodrugs/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Glioma/pathology , Haloperidol/chemical synthesis , Haloperidol/metabolism , Molecular Structure , Prodrugs/chemical synthesis , Prodrugs/metabolism , Rats , Structure-Activity Relationship , Tumor Cells, Cultured
11.
Eur J Med Chem ; 60: 285-94, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23313637

ABSTRACT

Phosphodiesterase type 5 (PDE5) mediates the degradation of cGMP in a variety of tissues including brain. Recent studies have demonstrated the importance of the nitric oxide/cGMP/cAMP-responsive element-binding protein (CREB) pathway to the process of learning and memory. Thus, PDE5 inhibitors (PDE5Is) are thought to be promising new therapeutic agents for the treatment of Alzheimer's disease (AD), a neurodegenerative disorder characterized by memory loss. To explore this possibility, a series of quinoline derivatives were synthesized and evaluated. We found that compound 7a selectively inhibits PDE5 with an IC(50) of 0.27 nM and readily crosses the blood brain barrier. In an in vivo mouse model of AD, compound 7a rescues synaptic and memory defects. Quinoline-based, CNS-permeant PDE5Is have potential for AD therapeutic development.


Subject(s)
Alzheimer Disease/drug therapy , Drug Discovery , Phosphodiesterase 5 Inhibitors/therapeutic use , Quinolines/therapeutic use , Animals , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Phosphodiesterase 5 Inhibitors/chemical synthesis , Phosphodiesterase 5 Inhibitors/chemistry , Quinolines/chemical synthesis , Quinolines/chemistry
12.
Eur J Med Chem ; 46(1): 433-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21055848

ABSTRACT

Complex mechanisms of prostate cancer progression prompt to novel therapeutic strategies concerning a combination of drugs or of single molecules able to interact with more crucial targets. Histone deacetylase inhibitors and sigma ligands with mixed σ(1) antagonist and σ(2) agonist properties were proposed as new potential tools for treatment of prostate cancer. (±)-MRJF4 was synthesized as phenylbutyrate ester of haloperidol metabolite II, which is a molecule consisting of a histone deacetilase inhibitor (4-phenylbutyric acid) and a sigma ligand (haloperidol metabolite II). Antiproliferatives activities of 4-phenylbutyric acid, haloperidol metabolite II, equimolar mixture of both compounds and (±)-MRJF4 were evaluated in vitro on LNCaP and PC3 prostate cancer cells. Preliminary binding studies of (±)-MRJF4 for σ(1), σ(2), D(2) and D(3) receptors and inhibition HDAC activity were reported. MTT cell viability assays highlighted a notable increase of antiproliferative activity of (±)-MRJF4 (IC(50) = 11 and 13 µM for LNCaP and PC3, respectively) compared to 4-phenylbutyric acid, haloperidol metabolite II and the respective equimolar pharmacological association. (±)-MRJF4 was also used in combination with σ(1) agonist (+)-pentazocine and σ(2) antagonist AC927 in order to evaluate the role of σ receptor subtypes in prostate cancer cell death.


Subject(s)
Haloperidol/analogs & derivatives , Haloperidol/metabolism , Phenylbutyrates/chemistry , Phenylbutyrates/pharmacology , Prostatic Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Guinea Pigs , Haloperidol/chemistry , Haloperidol/pharmacology , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Male , Phenylbutyrates/metabolism , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/metabolism , Receptors, sigma/agonists , Receptors, sigma/antagonists & inhibitors , Receptors, sigma/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL