Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Neurotherapeutics ; : e00372, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38760316

ABSTRACT

Spinal cord injury (SCI) is a life-changing event that severely impacts the patient's quality of life. Modulating neuroinflammation, which exacerbates the primary injury, and stimulating neuro-regenerative repair mechanisms are key strategies to improve functional recovery. Cyclic adenosine monophosphate (cAMP) is a second messenger crucially involved in both processes. Following SCI, intracellular levels of cAMP are known to decrease over time. Therefore, preventing cAMP degradation represents a promising strategy to suppress inflammation while stimulating regeneration. Intracellular cAMP levels are controlled by its hydrolyzing enzymes phosphodiesterases (PDEs). The PDE4 family is most abundantly expressed in the central nervous system (CNS) and its inhibition has been shown to be therapeutically relevant for managing SCI pathology. Unfortunately, the use of full PDE4 inhibitors at therapeutic doses is associated with severe emetic side effects, hampering their translation toward clinical applications. Therefore, in this study, we evaluated the effect of inhibiting specific PDE4 subtypes (PDE4B and PDE4D) on inflammatory and regenerative processes following SCI, as inhibitors selective for these subtypes have been demonstrated to be well-tolerated. We reveal that administration of the PDE4D inhibitor Gebr32a, even when starting 2 dpi, but not the PDE4B inhibitor A33, improved functional as well as histopathological outcomes after SCI, comparable to results obtained with the full PDE4 inhibitor roflumilast. Furthermore, using a luminescent human iPSC-derived neurospheroid model, we show that PDE4D inhibition stabilizes neural viability by preventing apoptosis and stimulating neuronal differentiation. These findings strongly suggest that specific PDE4D inhibition offers a novel therapeutic approach for SCI.

2.
Cell Mol Life Sci ; 80(7): 178, 2023 Jun 12.
Article in English | MEDLINE | ID: mdl-37306762

ABSTRACT

Inhibition of phosphodiesterase 4D (PDE4D) enzymes has been investigated as therapeutic strategy to treat memory problems in Alzheimer's disease (AD). Although PDE4D inhibitors are effective in enhancing memory processes in rodents and humans, severe side effects may hamper their clinical use. PDE4D enzymes comprise different isoforms, which, when targeted specifically, can increase treatment efficacy and safety. The function of PDE4D isoforms in AD and in molecular memory processes per se has remained unresolved. Here, we report the upregulation of specific PDE4D isoforms in transgenic AD mice and hippocampal neurons exposed to amyloid-ß. Furthermore, by means of pharmacological inhibition and CRISPR-Cas9 knockdown, we show that the long-form PDE4D3, -D5, -D7, and -D9 isoforms regulate neuronal plasticity and convey resilience against amyloid-ß in vitro. These results indicate that isoform-specific, next to non-selective, PDE4D inhibition is efficient in promoting neuroplasticity in an AD context. Therapeutic effects of non-selective PDE4D inhibitors are likely achieved through actions on long isoforms. Future research should identify which long PDE4D isoforms should be specifically targeted in vivo to both improve treatment efficacy and reduce side effects.


Subject(s)
Alzheimer Disease , Phosphoric Diester Hydrolases , Humans , Animals , Mice , Neurites , Amyloid beta-Peptides , Neurons , Mice, Transgenic , Cyclic Nucleotide Phosphodiesterases, Type 4
4.
Brain Behav Immun ; 109: 1-22, 2023 03.
Article in English | MEDLINE | ID: mdl-36584795

ABSTRACT

Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) characterized by focal inflammatory lesions and prominent demyelination. Even though the currently available therapies are effective in treating the initial stages of disease, they are unable to halt or reverse disease progression into the chronic progressive stage. Thus far, no repair-inducing treatments are available for progressive MS patients. Hence, there is an urgent need for the development of new therapeutic strategies either targeting the destructive immunological demyelination or boosting endogenous repair mechanisms. Using in vitro, ex vivo, and in vivo models, we demonstrate that selective inhibition of phosphodiesterase 4 (PDE4), a family of enzymes that hydrolyzes and inactivates cyclic adenosine monophosphate (cAMP), reduces inflammation and promotes myelin repair. More specifically, we segregated the myelination-promoting and anti-inflammatory effects into a PDE4D- and PDE4B-dependent process respectively. We show that inhibition of PDE4D boosts oligodendrocyte progenitor cells (OPC) differentiation and enhances (re)myelination of both murine OPCs and human iPSC-derived OPCs. In addition, PDE4D inhibition promotes in vivo remyelination in the cuprizone model, which is accompanied by improved spatial memory and reduced visual evoked potential latency times. We further identified that PDE4B-specific inhibition exerts anti-inflammatory effects since it lowers in vitro monocytic nitric oxide (NO) production and improves in vivo neurological scores during the early phase of experimental autoimmune encephalomyelitis (EAE). In contrast to the pan PDE4 inhibitor roflumilast, the therapeutic dose of both the PDE4B-specific inhibitor A33 and the PDE4D-specific inhibitor Gebr32a did not trigger emesis-like side effects in rodents. Finally, we report distinct PDE4D isoform expression patterns in human area postrema neurons and human oligodendroglia lineage cells. Using the CRISPR-Cas9 system, we confirmed that pde4d1/2 and pde4d6 are the key targets to induce OPC differentiation. Collectively, these data demonstrate that gene specific PDE4 inhibitors have potential as novel therapeutic agents for targeting the distinct disease processes of MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Phosphodiesterase 4 Inhibitors , Humans , Mice , Animals , Myelin Sheath/metabolism , Multiple Sclerosis/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4/therapeutic use , Evoked Potentials, Visual , Oligodendroglia/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Cell Differentiation , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/therapeutic use , Anti-Inflammatory Agents/pharmacology , Mice, Inbred C57BL
5.
Biomolecules ; 11(12)2021 11 24.
Article in English | MEDLINE | ID: mdl-34944403

ABSTRACT

The pyrazolyl-urea Gege3 molecule has shown interesting antiangiogenic effects in the tumor contest. Here, we have studied the role of this compound as interfering with endothelial cells activation in response to the paracrine effects of annexin A1 (ANXA1), known to be involved in promoting tumor progression. ANXA1 has been analyzed in the extracellular environment once secreted through microvesicles (EVs) by pancreatic cancer (PC) cells. Particularly, Gege3 has been able to notably prevent the effects of Ac2-26, the ANXA1 mimetic peptide, and of PC-derived EVs on endothelial cells motility, angiogenesis, and calcium release. Furthermore, this compound also inhibited the translocation of ANXA1 to the plasma membrane, otherwise induced by the same ANXA1-dependent extracellular stimuli. Moreover, these effects have been mediated by the indirect inhibition of protein kinase Cα (PKCα), which generally promotes the phosphorylation of ANXA1 on serine 27. Indeed, by the subtraction of intracellular calcium levels, the pathway triggered by PKCα underwent a strong inhibition leading to the following impediment to the ANXA1 localization at the plasma membrane, as revealed by confocal and cytofluorimetry analysis. Thus, Gege3 appeared an attractive molecule able to prevent the paracrine effects of PC cells deriving ANXA1 in the tumor microenvironment.


Subject(s)
Annexin A1/metabolism , Down-Regulation , Extracellular Vesicles/metabolism , Pancreatic Neoplasms/metabolism , Small Molecule Libraries/pharmacology , Urea/chemistry , Annexin A1/pharmacology , Calcium/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement/drug effects , Extracellular Vesicles/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Pancreatic Neoplasms/drug therapy , Paracrine Communication/drug effects , Peptides/pharmacology , Phosphorylation/drug effects , Protein Transport/drug effects , Small Molecule Libraries/chemistry , Tumor Microenvironment/drug effects
6.
Eur J Med Chem ; 226: 113872, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34600191

ABSTRACT

Based on biological results of previous synthesized pyrazolyl ureas able to interfere with angiogenesis process, we planned and synthesized the new benzyl-urea derivatives 2-4; some of them showed an interesting anti-proliferative profile and particularly 4e potently inhibited HUVEC proliferation. To shed light on the mechanism of action of 4e, its interactome has been deeply inspected to identify the most prominent protein partners, mainly taking into account kinome and phosphatome, through drug affinity responsive target stability experiments, followed by targeted limited proteolysis analysis. From these studies, PP1γ emerged as the most reliable 4e potential target in HUVEC. Molecular docking simulations on PP1γ were carried out to predict 4e binding mode. To assess its potential anti-angiogenic effect, 4e was tested in vitro to verify interference on kinase and phosphate activities. Overall, our results evidenced for 4e an interesting anti-angiogenic action, probably due to its action at intracellular level on PP1γ signalling pathways.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Neovascularization, Physiologic/drug effects , Pyrazoles/pharmacology , Urea/pharmacology , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Structure , Protein Phosphatase 1/antagonists & inhibitors , Protein Phosphatase 1/metabolism , Proteolysis/drug effects , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Structure-Activity Relationship , Urea/analogs & derivatives , Urea/chemistry
7.
Molecules ; 26(19)2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34641279

ABSTRACT

(1) Background: different previously synthesized pyrazoles and imidazo-pyrazoles showed interesting anti-angiogenic action, being able to interfere with ERK1/2, AKT and p38MAPK phosphorylation in different manners and with different potency; (2) Methods: here, a new small compound library, endowed with the same differently decorated chemical scaffolds, has been synthetized to obtain new agents able to inhibit different pathways involved in inflammation, cancer and human platelet aggregation. (3) Results: most of the new synthesized derivatives resulted able to block ROS production, platelet aggregation and p38MAPK phosphorylation both in platelets and Human Umbilical Vein Endothelial cells (HUVEC). This paves the way for the development of new agents with anti-angiogenic activity.


Subject(s)
Anti-Inflammatory Agents/chemical synthesis , Imidazoles/chemistry , Pyrazoles/chemistry , Small Molecule Libraries/chemical synthesis , p38 Mitogen-Activated Protein Kinases/metabolism , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Phosphorylation/drug effects , Platelet Aggregation/drug effects , Reactive Oxygen Species/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
8.
Bioorg Chem ; 115: 105168, 2021 10.
Article in English | MEDLINE | ID: mdl-34284173

ABSTRACT

In recent years, 5-pyrazolyl-ureas have mostly been known for their attractive poly-pharmacological outline and, in particular, ethyl 1-(2-hydroxypentyl)-5-(3-(3-(trifluoromethyl) phenyl) ureido)-1H-pyrazole-4-carboxylate (named GeGe-3) has emerged as a capable anti-angiogenic compound. This paper examines its interactome by functional proteomics using a label-free mass spectrometry based platform, coupling Drug Affinity Responsive Target Stability and targeted Limited Proteolysis-Multiple Reaction Monitoring. Calreticulin has been recognized as the GeGe-3 principal target and this evidence has been supported by immunoblotting and in silico molecular docking. Furthermore, cell studies have shown that GeGe-3 lowers cell calcium mobilization, cytoskeleton organization and focal adhesion kinase expression, thus linking its biological potential to calreticulin binding and, ultimately, shedding light on the reasonable action mechanism of this molecule as an anti-angiogenic factor.


Subject(s)
Angiogenesis Inhibitors/chemistry , Proteome/metabolism , Proteomics/methods , Pyrazoles/chemistry , Urea/chemistry , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/pharmacology , Binding Sites , Calcium/metabolism , Calreticulin/chemistry , Calreticulin/metabolism , Down-Regulation/drug effects , Focal Adhesion Kinase 1/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Ionomycin/pharmacology , Molecular Docking Simulation , Proteome/drug effects
9.
Eur J Med Chem ; 223: 113638, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34171658

ABSTRACT

Memory and cognitive functions depend on the cerebral levels of cyclic adenosine monophosphate (cAMP), which are regulated by the phosphodiesterase 4 (PDE4) family of enzymes. Selected rolipram-related PDE4 inhibitors, members of the GEBR library, have been shown to increase hippocampal cAMP levels, providing pro-cognitive benefits with a safe pharmacological profile. In a recent SAR investigation involving a subset of GEBR library compounds, we have demonstrated that, depending on length and flexibility, ligands can either adopt a twisted, an extended or a protruding conformation, the latter allowing the ligand to form stabilizing contacts with the regulatory domain of the enzyme. Here, based on those findings, we describe further chemical modifications of the protruding subset of GEBR library inhibitors and their effects on ligand conformation and potency. In particular, we demonstrate that the insertion of a methyl group in the flexible linker region connecting the catechol portion and the basic end of the molecules enhances the ability of the ligand to interact with both the catalytic and the regulatory domains of the enzyme.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Drug Design , Phosphodiesterase 4 Inhibitors/chemical synthesis , Small Molecule Libraries/chemistry , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Humans , Molecular Docking Simulation , Phosphodiesterase 4 Inhibitors/chemistry , Phosphodiesterase 4 Inhibitors/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/metabolism , Structure-Activity Relationship
10.
Oncotarget ; 11(37): 3459-3472, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32973970

ABSTRACT

Pyrazolyl-urea and dihydro-imidazo-pyrazolyl-urea compounds (STIRUR 13, STIRUR 41 and BUR 12) have been demonstrated to exert a strong inhibitory effect on interleukin 8 or N-formyl-methionyl-leucyl-phenylalanine-induced chemotaxis of human neutrophils. Since the migration of cancer cells is comparable to that of neutrophils, the purpose of this study is to evaluate the biological effect of STIRUR 13, STIRUR 41 and BUR 12 on ACN and HTLA-230, two neuroblastoma (NB) cell lines with different degree of malignancy. HTLA-230 cells, stage-IV NB cells, have high plasticity and can serve as progenitors of endothelial cells. The results herein reported show that the three tested compounds were not cytotoxic for both NB cells and did not alter their clonogenic potential. However, all compounds were able to inhibit the ability of HTLA-230 to form vascular-like structures. On the basis of these findings, pyrazolyl-urea and dihydro-imidazo-pyrazolyl-urea derivatives could be proposed as agents potentially effective in counteracting NB malignancy by inhibiting cell migration and tumor angiogenesis which represent important hallmarks responsible for cancer survival and progression.

11.
Molecules ; 25(15)2020 Jul 29.
Article in English | MEDLINE | ID: mdl-32751358

ABSTRACT

The pyrazole nucleus has long been known as a privileged scaffold in the synthesis of biologically active compounds. Within the numerous pyrazole derivatives developed as potential drugs, this review is focused on molecules characterized by a urea function directly linked to the pyrazole nucleus in a different position. In the last 20 years, the interest of numerous researchers has been especially attracted by pyrazolyl-ureas showing a wide spectrum of biological activities, ranging from the antipathogenic activities (bacteria, plasmodium, toxoplasma, and others) to the anticarcinogenic activities. In particular, in the anticancer field, pyrazolyl-ureas have been shown to interact at the intracellular level on many pathways, in particular on different kinases such as Src, p38-MAPK, TrKa, and others. In addition, some of them evidenced an antiangiogenic potential that deserves to be explored. This review therefore summarizes all these biological data (from 2000 to date), including patented compounds.


Subject(s)
Chemistry, Pharmaceutical , Pharmaceutical Preparations/chemistry , Pyrazoles/chemistry , Urea/chemistry , Chemistry Techniques, Synthetic , Chemistry, Pharmaceutical/methods , Humans , Pharmaceutical Preparations/chemical synthesis , Structure-Activity Relationship
12.
ChemMedChem ; 15(11): 961-969, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32233110

ABSTRACT

Even though immunotherapy has radically changed the search for anticancer therapies, there are still many different pathways that are open to intervention with traditional small molecules. To expand our investigation in the anticancer field, we report here a new series of compounds in which our previous pyrazole and imidazopyrazole scaffolds are linked to a differently decorated phenyl ring through an acylhydrazone linker. Preliminary tests on the library were performed at the National Cancer Institute (USA) against the full NCI 60 cell panel. The best compounds among the imidazopyrazole series were then tested by immunofluorescence staining for their inhibition of cell proliferation, apoptosis induction, and their effect on the cell cycle and on microtubules. Two compounds, in particular 4-benzyloxy-3-methoxybenzyliden imidazopyrazole-7-carbohydrazide showed good growth inhibition, with IC50 values in the low-micromolar range, and induced apoptosis. Both compounds altered the cell-cycle phases with the appearance of polyploid cells. Immunofluorescence analysis evidenced microtubules alterations; tubulin polymerization assays and docking studies suggested the tubulin system to be the possible, although not exclusive, target of the new acylhydrazone series reported here.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Hydrazones/pharmacology , Microtubules/drug effects , Pyrazoles/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Hydrazones/chemical synthesis , Hydrazones/chemistry , Molecular Structure , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
13.
Molecules ; 25(4)2020 Feb 19.
Article in English | MEDLINE | ID: mdl-32093112

ABSTRACT

Alzheimer's disease is the most common type of dementia, affecting millions of people worldwide. One of its main consequences is memory loss, which is related to downstream effectors of cyclic adenosine monophosphate (cAMP). A well-established strategy to avoid cAMP degradation is the inhibition of phosphodiesterase (PDE). In recent years, GEBR-32a has been shown to possess selective inhibitory properties against PDE type 4 family members, resulting in an improvement in spatial memory processes without the typical side effects that are usually correlated with this mechanism of action. In this work, we performed the HPLC chiral resolution and absolute configuration assignment of GEBR-32a. We developed an efficient analytical and semipreparative chromatographic method exploiting an amylose-based stationary phase, we studied the chiroptical properties of both enantiomers and we assigned their absolute configuration by 1H-NMR (nuclear magnetic resonance). Lastly, we measured the IC50 values of both enantiomers against both the PDE4D catalytic domain and the long PDE4D3 isoform. Results strongly support the notion that GEBR-32a inhibits the PDE4D enzyme by interacting with both the catalytic pocket and the regulatory domains.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Phosphodiesterase 4 Inhibitors/chemistry , Humans , Nuclear Magnetic Resonance, Biomolecular
14.
Molecules ; 25(4)2020 Feb 18.
Article in English | MEDLINE | ID: mdl-32085423

ABSTRACT

Several anti-inflammatory agents based on pyrazole and imidazopyrazole scaffolds and a large library of substituted catechol PDE4D inhibitors were reported by us in the recent past. To obtain new molecules potentially able to act on different targets involved in inflammation onset we designed and synthesized a series of hybrid compounds by linking pyrazole and imidazo-pyrazole scaffolds to differently decorated catechol moieties through an acylhydrazone chain. Some compounds showed antioxidant activity, inhibiting reactive oxygen species (ROS) elevation in neutrophils, and a good inhibition of phosphodiesterases type 4D and, particularly, type 4B, the isoform most involved in inflammation. In addition, most compounds inhibited ROS production also in platelets, confirming their ability to exert an antiinflammatory response by two independent mechanism. Structure-activity relationship (SAR) analyses evidenced that both heterocyclic scaffolds (pyrazole and imidazopyrazole) and the substituted catechol moiety were determinant for the pharmacodynamic properties, even if hybrid molecules bearing to the pyrazole series were more active than the imidazopyrazole ones. In addition, the pivotal role of the catechol substituents has been analyzed. In conclusion the hybridization approach gave a new serie of multitarget antiinflammatory compounds, characterized by a strong antioxidant activity in different biological targets.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Pyrazoles/pharmacology , Reactive Oxygen Species/metabolism , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/chemistry , Blood Platelets/drug effects , Blood Platelets/metabolism , Cell Survival/drug effects , Chemotaxis/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/pharmacology , Humans , Male , Neutrophils/drug effects , Neutrophils/metabolism , Oxidation-Reduction , Phosphodiesterase 4 Inhibitors/chemical synthesis , Phosphodiesterase 4 Inhibitors/pharmacology , Platelet Aggregation/drug effects , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Structure-Activity Relationship
15.
Med Chem ; 15(1): 17-27, 2019.
Article in English | MEDLINE | ID: mdl-29792151

ABSTRACT

BACKGROUND: We screened a large library of differently decorated imidazo-pyrazole and pyrazole derivatives as possible new antitubercular agents and this preliminary screening showed that many compounds are able to totally inhibit Mycobacterium growth (>90 %). Among the most active compounds, we selected some new possible hits based on their similarities and, at the same time, on their novelty with respect to the pipeline drugs. METHODS: In order to increase the potency and obtain more information about structure-activity relationship (SAR), we designed and synthesized three new series of compounds (2a-e, 3a-e, and 4a-l). CONCLUSION: Performed tests confirmed that both new pyrazoles and imidazo-pyrazoles could represent a new starting point to obtain more potent compounds and further work is now underway to identify the protein targets of this new class of anti-TB agents.


Subject(s)
Antitubercular Agents/pharmacology , Imidazoles/pharmacology , Pyrazoles/pharmacology , Small Molecule Libraries/pharmacology , Animals , Antitubercular Agents/chemical synthesis , Antitubercular Agents/chemistry , Antitubercular Agents/toxicity , Chlorocebus aethiops , Imidazoles/chemical synthesis , Imidazoles/chemistry , Imidazoles/toxicity , Microbial Sensitivity Tests , Mycobacterium tuberculosis/drug effects , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrazoles/toxicity , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Vero Cells
16.
Biochemistry ; 57(19): 2876-2888, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29652483

ABSTRACT

Selected members of the large rolipram-related GEBR family of type 4 phosphodiesterase (PDE4) inhibitors have been shown to facilitate long-term potentiation and to improve memory functions without causing emetic-like behavior in rodents. Despite their micromolar-range binding affinities and their promising pharmacological and toxicological profiles, few if any structure-activity relationship studies have been performed to elucidate the molecular bases of their action. Here, we report the crystal structure of a number of GEBR library compounds in complex with the catalytic domain of PDE4D as well as their inhibitory profiles for both the long PDE4D3 isoform and the catalytic domain alone. Furthermore, we assessed the stability of the observed ligand conformations in the context of the intact enzyme using molecular dynamics simulations. The longer and more flexible ligands appear to be capable of forming contacts with the regulatory portion of the enzyme, thus possibly allowing some degree of selectivity between the different PDE4 isoforms.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Memory/drug effects , Phosphodiesterase 4 Inhibitors/chemistry , Structure-Activity Relationship , Animals , Catalytic Domain , Crystallography, X-Ray , Humans , Ligands , Memory/physiology , Molecular Dynamics Simulation , Phosphodiesterase 4 Inhibitors/therapeutic use , Rolipram/chemistry , Rolipram/therapeutic use
17.
Eur J Med Chem ; 133: 24-35, 2017 Jun 16.
Article in English | MEDLINE | ID: mdl-28371678

ABSTRACT

Taking into account the structure activity relationship information given by our previous studies, we designed and synthesized a small library of pyrazolylureas and imidazopyrazolecarboxamides fluorinated on urea moiety and differently decorated on pyrazole nucleus. All compounds were preliminary screened by Western blotting technique to evaluate their activity on MAPK and PI3K pathways by monitoring ERK1/2, p38MAPK and Akt phosphorylation, and also screened with a wound healing assay to assess their capacity in inhibiting endothelial cell migration, using human umbilical vein endothelial cells stimulated with VEGF. Pyrazoles and imidazopyrazoles did not show the same activity profile. SAR consideration showed that specific substituents and their position in pyrazole nucleus, as well as the type of substituent on the phenylurea moiety play a pivotal role in determining increase or decrease of kinases phosphorylation. On the other hand the loss of flexibility in imidazopyrazole derivatives is responsible for activity potentiation. Screening of the compound library for inhibition of endothelial cell migration, a function required for angiogenesis, showed significant activity for compound 3. This compound might interfere with cell migration by modulating the activity of different upstream target kinases. Therefore, compound 3 represents a potential inhibitor of angiogenesis. Furthermore, it may be used as a tool to identify unknown mediators of endothelial migration and thereby unveiling new therapeutic targets for controlling pathological angiogenesis in diseases such as cancers.


Subject(s)
Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Urea/analogs & derivatives , Urea/pharmacology , Cell Movement/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Phosphorylation/drug effects , Vascular Endothelial Growth Factor A/metabolism
18.
Sci Rep ; 7: 46320, 2017 04 12.
Article in English | MEDLINE | ID: mdl-28402318

ABSTRACT

Memory loss characterizes several neurodegenerative disorders, including Alzheimer's disease (AD). Inhibition of type 4 phosphodiesterase (PDE4) and elevation of cyclic adenosine monophosphate (cAMP) has emerged as a promising therapeutic approach to treat cognitive deficits. However, PDE4 exists in several isoforms and pan inhibitors cannot be used in humans due to severe emesis. Here, we present GEBR-32a, a new PDE4D full inhibitor that has been characterized both in vitro and in vivo using biochemical, electrophysiological and behavioural analyses. GEBR-32a efficiently enhances cAMP in neuronal cultures and hippocampal slices. In vivo pharmacokinetic analysis shows that GEBR-32a is rapidly distributed within the central nervous system with a very favourable brain/blood ratio. Specific behavioural tests (object location and Y-maze continuous alternation tasks) demonstrate that this PDE4D inhibitor is able to enhance memory in AD transgenic mice and concomitantly rescues their hippocampal long-term potentiation deficit. Of great relevance, our preliminary toxicological analysis indicates that GEBR-32a is not cytotoxic and genotoxic, and does not seem to possess emetic-like side effects. In conclusion, GEBR-32a could represent a very promising cognitive-enhancing drug with a great potential for the treatment of Alzheimer's disease.


Subject(s)
Memory/drug effects , Phosphodiesterase 4 Inhibitors/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Animals , Cells, Cultured , Cyclic AMP , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , DNA Damage/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Intracellular Space , Isoenzymes/antagonists & inhibitors , Long-Term Potentiation/drug effects , Mice , Mice, Transgenic , Molecular Structure , Phosphodiesterase 4 Inhibitors/chemical synthesis , Recombinant Proteins
19.
J Enzyme Inhib Med Chem ; 32(1): 375-402, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28276287

ABSTRACT

Respiratory syncytial virus (RSV) has been identified as a main cause of hospitalisation in infants and children. To date, the current therapeutic arsenal is limited to ribavirin and palivizumab with variable efficacy. In this work, starting from a number of in-house series of previously described anti-RSV agents based on the benzimidazole scaffold, with the aim at gaining a better understanding of the related chemical features involved in potency and safety profiles, we applied a computational study including two focussed comparative molecular fields analysis (CoMFA) and comparative molecular similarity indices analysis (CoMSIA). The results allowed us to derive useful suggestions for the design of derivatives and also to set up statistical models predicting the potency and selectivity index (SI = CC50/EC50) of any new analogue prior to synthesis. Accordingly, here, we discuss preliminary results obtained through the applied exhaustive QSAR analyses, leading to design and synthesise more effective anti-RSV agents.


Subject(s)
Antiviral Agents/pharmacology , Benzimidazoles/pharmacology , Respiratory Syncytial Viruses/drug effects , Respirovirus Infections/drug therapy , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Benzimidazoles/chemical synthesis , Benzimidazoles/chemistry , Cell Line , Chlorocebus aethiops , Dose-Response Relationship, Drug , Humans , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Quantitative Structure-Activity Relationship , Vero Cells
20.
Bioorg Med Chem Lett ; 27(1): 24-29, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27890378

ABSTRACT

Phosphodiesterase 4 (PDE4) inhibitors have attractive therapeutic potential in respiratory, inflammatory, metabolic and CNS disorders. The present work details the design, chemical exploration and biological profile of a novel PDE4 inhibitor chemotype. A diazepinone ring was identified as an under-represented heterocyclic system fulfilling a set of PDE4 structure-based design hypotheses. Rapid exploration of the structure activity relationships for the series was enabled by robust and scalable two/three-steps parallel chemistry protocols. The resulting compounds demonstrated PDE4 inhibitory activity in cell free and cell-based assays comparable to the Zardaverine control used, suggesting potential avenues for their further development.


Subject(s)
Azepines/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Drug Design , Phosphodiesterase 4 Inhibitors/pharmacology , Azepines/chemical synthesis , Azepines/chemistry , Dose-Response Relationship, Drug , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Molecular Structure , Phosphodiesterase 4 Inhibitors/chemical synthesis , Phosphodiesterase 4 Inhibitors/chemistry , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...