Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters

Publication year range
1.
Brain ; 145(1): 76-82, 2022 03 29.
Article in English | MEDLINE | ID: mdl-34196695

ABSTRACT

Fragile X syndrome is the most common inherited intellectual disability and mono-genetic cause of autism spectrum disorder. It is a neurodevelopmental condition occurring due to a CGG trinucleotide expansion in the FMR1 gene. Polymorphisms and variants in large-conductance calcium-activated potassium channels are increasingly linked to intellectual disability and loss of FMR protein causes reduced large-conductance calcium-activated potassium channel activity leading to abnormalities in synapse function. Using the cannabinoid-like large-conductance calcium-activated potassium channel activator VSN16R we rescued behavioural deficits such as repetitive behaviour, hippocampal dependent tests of daily living, hyperactivity and memory in a mouse model of fragile X syndrome. VSN16R has been shown to be safe in a phase 1 study in healthy volunteers and in a phase 2 study in patients with multiple sclerosis with high oral bioavailability and no serious adverse effects reported. VSN16R could therefore be directly utilized in a fragile X syndrome clinical study. Moreover, VSN16R showed no evidence of tolerance, which strongly suggests that chronic VSN16R may have great therapeutic value for fragile X syndrome and autism spectrum disorder. This study provides new insight into the pathophysiology of fragile X syndrome and identifies a new pathway for drug intervention for this debilitating disorder.


Subject(s)
Autism Spectrum Disorder , Cannabinoids , Fragile X Syndrome , Animals , Cannabinoids/pharmacology , Cannabinoids/therapeutic use , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/drug therapy , Fragile X Syndrome/genetics , Humans , Mice , Phenotype
2.
Chembiochem ; 23(1): e202100463, 2022 01 05.
Article in English | MEDLINE | ID: mdl-34647407

ABSTRACT

Vascular endothelial growth factors (VEGFs) regulate significant pathways in angiogenesis, myocardial and neuronal protection, metabolism, and cancer progression. The VEGF-B growth factor is involved in cell survival, anti-apoptotic and antioxidant mechanisms, through binding to VEGF receptor 1 and neuropilin-1 (NRP1). We employed surface plasmon resonance technology and X-ray crystallography to analyse the molecular basis of the interaction between VEGF-B and the b1 domain of NRP1, and developed VEGF-B C-terminus derived peptides to be used as chemical tools for studying VEGF-B - NRP1 related pathways. Peptide lipidation was used as a means to stabilise the peptides. VEGF-B-derived peptides containing a C-terminal arginine show potent binding to NRP1-b1. Peptide lipidation increased binding residence time and improved plasma stability. A crystal structure of a peptide with NRP1 demonstrated that VEGF-B peptides bind at the canonical C-terminal arginine binding site. VEGF-B C-terminus imparts higher affinity for NRP1 than the corresponding VEGF-A165 region. This tight binding may impact on the activity and selectivity of the full-length protein. The VEGF-B167 derived peptides were more effective than VEGF-A165 peptides in blocking functional phosphorylation events. Blockers of VEGF-B function have potential applications in diabetes and non-alcoholic fatty liver disease.


Subject(s)
Neuropilin-1/metabolism , Peptides/metabolism , Vascular Endothelial Growth Factor B/metabolism , Humans , Neuropilin-1/chemistry , Peptides/chemistry , Protein Binding , Vascular Endothelial Growth Factor B/chemistry
3.
EMBO Rep ; 20(11): e47967, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31566294

ABSTRACT

Dystroglycan, an extracellular matrix receptor, has essential functions in various tissues. Loss of α-dystroglycan-laminin interaction due to defective glycosylation of α-dystroglycan underlies a group of congenital muscular dystrophies often associated with brain malformations, referred to as dystroglycanopathies. The lack of isogenic human dystroglycanopathy cell models has limited our ability to test potential drugs in a human- and neural-specific context. Here, we generated induced pluripotent stem cells (iPSCs) from a severe dystroglycanopathy patient with homozygous FKRP (fukutin-related protein gene) mutation. We showed that CRISPR/Cas9-mediated gene correction of FKRP restored glycosylation of α-dystroglycan in iPSC-derived cortical neurons, whereas targeted gene mutation of FKRP in wild-type cells disrupted this glycosylation. In parallel, we screened 31,954 small molecule compounds using a mouse myoblast line for increased glycosylation of α-dystroglycan. Using human FKRP-iPSC-derived neural cells for hit validation, we demonstrated that compound 4-(4-bromophenyl)-6-ethylsulfanyl-2-oxo-3,4-dihydro-1H-pyridine-5-carbonitrile (4BPPNit) significantly augmented glycosylation of α-dystroglycan, in part through upregulation of LARGE1 glycosyltransferase gene expression. Together, isogenic human iPSC-derived cells represent a valuable platform for facilitating dystroglycanopathy drug discovery and therapeutic development.


Subject(s)
Drug Evaluation, Preclinical , Dystroglycans/metabolism , Induced Pluripotent Stem Cells/metabolism , Base Sequence , CRISPR-Cas Systems , Cells, Cultured , Drug Evaluation, Preclinical/methods , Dystroglycans/genetics , Gene Editing , Gene Targeting , Genetic Loci , Glycosylation/drug effects , High-Throughput Nucleotide Sequencing , Humans , Molecular Imaging , Muscular Dystrophies/drug therapy , Muscular Dystrophies/etiology , Muscular Dystrophies/metabolism , Mutation , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Neural Stem Cells/metabolism , Neurons/metabolism , Pentosyltransferases/genetics , Pentosyltransferases/metabolism
4.
Drug Dev Res ; 81(4): 491-500, 2020 06.
Article in English | MEDLINE | ID: mdl-31958155

ABSTRACT

Neuropilin-1 (NRP1) is emerging as an important molecule in immune signaling where it has been shown to modulate the actions of TGF-ß1 in macrophages and regulatory T cells. The development of cost-effective and reliable assays for NRP1 binding is therefore important. We synthesized three new NRP1 small molecule fluorophores and examined their performance as fluorescent polarization probes. One molecule DS108 exhibited favorable binding and fluorescent characteristics and allowed us to establish a simple assay suitable for medium to high throughput screening of small molecules.


Subject(s)
Fluorescent Dyes/metabolism , High-Throughput Screening Assays/methods , Neuropilin-1/metabolism , Fluorescent Dyes/chemical synthesis , Signal Transduction , Transforming Growth Factor beta1/metabolism
5.
Nature ; 503(7476): 402-405, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24196705

ABSTRACT

Human immunodeficiency virus (HIV)-1 is able to replicate in primary human macrophages without stimulating innate immunity despite reverse transcription of genomic RNA into double-stranded DNA, an activity that might be expected to trigger innate pattern recognition receptors. We reasoned that if correctly orchestrated HIV-1 uncoating and nuclear entry is important for evasion of innate sensors then manipulation of specific interactions between HIV-1 capsid and host factors that putatively regulate these processes should trigger pattern recognition receptors and stimulate type 1 interferon (IFN) secretion. Here we show that HIV-1 capsid mutants N74D and P90A, which are impaired for interaction with cofactors cleavage and polyadenylation specificity factor subunit 6 (CPSF6) and cyclophilins (Nup358 and CypA), respectively, cannot replicate in primary human monocyte-derived macrophages because they trigger innate sensors leading to nuclear translocation of NF-κB and IRF3, the production of soluble type 1 IFN and induction of an antiviral state. Depletion of CPSF6 with short hairpin RNA expression allows wild-type virus to trigger innate sensors and IFN production. In each case, suppressed replication is rescued by IFN-receptor blockade, demonstrating a role for IFN in restriction. IFN production is dependent on viral reverse transcription but not integration, indicating that a viral reverse transcription product comprises the HIV-1 pathogen-associated molecular pattern. Finally, we show that we can pharmacologically induce wild-type HIV-1 infection to stimulate IFN secretion and an antiviral state using a non-immunosuppressive cyclosporine analogue. We conclude that HIV-1 has evolved to use CPSF6 and cyclophilins to cloak its replication, allowing evasion of innate immune sensors and induction of a cell-autonomous innate immune response in primary human macrophages.


Subject(s)
HIV-1/immunology , Immune Evasion , Immunity, Innate/immunology , Macrophages/immunology , Macrophages/virology , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cyclophilins/metabolism , Cyclosporine/metabolism , HIV Infections/immunology , HIV Infections/metabolism , HIV Infections/pathology , HIV Infections/virology , HIV-1/metabolism , Humans , Interferon Regulatory Factor-3/metabolism , Interferon Type I/immunology , Interferon Type I/metabolism , Macrophages/cytology , Macrophages/pathology , Molecular Chaperones/metabolism , Monocytes/cytology , NF-kappa B/metabolism , Nuclear Pore Complex Proteins/metabolism , Receptors, Pattern Recognition , Virus Internalization , Virus Replication/immunology , mRNA Cleavage and Polyadenylation Factors/deficiency , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism
6.
J Virol ; 91(23)2017 12 01.
Article in English | MEDLINE | ID: mdl-28931678

ABSTRACT

Primary effusion lymphoma (PEL) is a lymphogenic disorder associated with Kaposi's sarcoma-associated herpesvirus (KSHV) infection. Key to the survival and proliferation of PEL is the canonical NF-κB pathway, which becomes constitutively activated following overexpression of the viral oncoprotein KSHV vFLIP (ks-vFLIP). This arises from its capacity to form a complex with the modulatory subunit of the IκB kinase (IKK) kinase, IKKγ (or NEMO), resulting in the overproduction of proteins that promote cellular survival and prevent apoptosis, both of which are important drivers of tumorigenesis. Using a combination of cell-based and biophysical assays together with structural techniques, we showed that the observed resistance to cell death is largely independent of autophagy or major death receptor signaling pathways and demonstrated that direct targeting of the ks-vFLIP-IKKγ interaction both in cells and in vitro can be achieved using IKKγ-mimetic peptides. Our results further reveal that these peptides not only induce cell killing but also potently sensitize PEL to the proapoptotic agents tumor necrosis factor alpha and etoposide and are the first to confirm ks-vFLIP as a tractable target for the treatment of PEL and related disorders.IMPORTANCE KSHV vFLIP (ks-vFLIP) has been shown to have a crucial role in cellular transformation, in which it is vital for the survival and proliferation of primary effusion lymphoma (PEL), an aggressive malignancy associated with infection that is resistant to the majority of chemotherapeutic drugs. It operates via subversion of the canonical NF-κB pathway, which requires a physical interaction between ks-vFLIP and the IKK kinase modulatory subunit IKKγ. While this interaction has been directly linked to protection against apoptosis, it is unclear whether the suppression of other cell death pathways implicated in ks-vFLIP pathogenesis is an additional contributor. We demonstrate that the interaction between ks-vFLIP and IKKγ is pivotal in conferring resistance to apoptosis. Additionally, we show that the ks-vFLIP-IKKγ complex can be disrupted using peptides leading to direct killing and the sensitization of PEL cells to proapoptotic agents. Our studies thus provide a framework for future therapeutic interventions.


Subject(s)
Apoptosis , Herpesvirus 8, Human/physiology , I-kappa B Kinase/chemistry , Peptides/metabolism , Peptides/pharmacology , Sarcoma, Kaposi/virology , Autophagy , Etoposide/pharmacology , Herpesvirus 8, Human/chemistry , Humans , I-kappa B Kinase/metabolism , Jurkat Cells , Molecular Mimicry , Peptides/chemistry , Protein Binding , Sarcoma, Kaposi/physiopathology , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Viral Proteins/metabolism
7.
J Biol Chem ; 291(9): 4356-73, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26679998

ABSTRACT

The mitochondrial permeability transition pore is a recognized drug target for neurodegenerative conditions such as multiple sclerosis and for ischemia-reperfusion injury in the brain and heart. The peptidylprolyl isomerase, cyclophilin D (CypD, PPIF), is a positive regulator of the pore, and genetic down-regulation or knock-out improves outcomes in disease models. Current inhibitors of peptidylprolyl isomerases show no selectivity between the tightly conserved cyclophilin paralogs and exhibit significant off-target effects, immunosuppression, and toxicity. We therefore designed and synthesized a new mitochondrially targeted CypD inhibitor, JW47, using a quinolinium cation tethered to cyclosporine. X-ray analysis was used to validate the design concept, and biological evaluation revealed selective cellular inhibition of CypD and the permeability transition pore with reduced cellular toxicity compared with cyclosporine. In an experimental autoimmune encephalomyelitis disease model of neurodegeneration in multiple sclerosis, JW47 demonstrated significant protection of axons and improved motor assessments with minimal immunosuppression. These findings suggest that selective CypD inhibition may represent a viable therapeutic strategy for MS and identify quinolinium as a mitochondrial targeting group for in vivo use.


Subject(s)
Cerebral Cortex/drug effects , Cyclophilins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Multiple Sclerosis/prevention & control , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Quinolinium Compounds/therapeutic use , Amino Acid Substitution , Animals , Cell Proliferation/drug effects , Cells, Cultured , Cerebral Cortex/immunology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Peptidyl-Prolyl Isomerase F , Cyclophilins/genetics , Cyclophilins/metabolism , Cyclosporins/adverse effects , Cyclosporins/chemical synthesis , Cyclosporins/pharmacology , Cyclosporins/therapeutic use , Hep G2 Cells , Humans , Liver/drug effects , Liver/metabolism , Male , Mice, Inbred Strains , Mice, Knockout , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Mutation , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/adverse effects , Neuroprotective Agents/pharmacology , Peptides, Cyclic/adverse effects , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/pharmacology , Peptides, Cyclic/therapeutic use , Quinolinium Compounds/adverse effects , Quinolinium Compounds/chemical synthesis , Quinolinium Compounds/pharmacology , Random Allocation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/pathology
8.
J Virol ; 90(19): 8552-62, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27440884

ABSTRACT

UNLABELLED: Tripartite motif-containing protein 5 (TRIM5) restricts human immunodeficiency virus type 1 (HIV-1) in a species-specific manner by uncoating viral particles while activating early innate responses. Although the contribution of TRIM5 proteins to cellular immunity has not yet been studied, their interactions with the incoming viral capsid and the cellular proteasome led us to hypothesize a role for them. Here, we investigate whether the expression of two nonhuman TRIM5 orthologs, rhesus TRIM5α (RhT5) and TRIM-cyclophilin A (TCyp), both of which are potent restrictors of HIV-1, could enhance immune recognition of infected cells by CD8(+) T cells. We illustrate how TRIM5 restriction improves CD8(+) T-cell-mediated HIV-1 inhibition. Moreover, when TRIM5 activity was blocked by the nonimmunosuppressive analog of cyclosporine (CsA), sarcosine-3(4-methylbenzoate)-CsA (SmBz-CsA), we found a significant reduction in CD107a/MIP-1ß expression in HIV-1-specific CD8(+) T cells. This finding underscores the direct link between TRIM5 restriction and activation of CD8(+) T-cell responses. Interestingly, cells expressing RhT5 induced stronger CD8(+) T-cell responses through the specific recognition of the HIV-1 capsid by the immune system. The underlying mechanism of this process may involve TRIM5-specific capsid recruitment to cellular proteasomes and increase peptide availability for loading and presentation of HLA class I antigens. In summary, we identified a novel function for nonhuman TRIM5 variants in cellular immunity. We hypothesize that TRIM5 can couple innate viral sensing and CD8(+) T-cell activation to increase species barriers against retrovirus infection. IMPORTANCE: New therapeutics to tackle HIV-1 infection should aim to combine rapid innate viral sensing and cellular immune recognition. Such strategies could prevent seeding of the viral reservoir and the immune damage that occurs during acute infection. The nonhuman TRIM5 variants, rhesus TRIM5α (RhT5) and TRIM-cyclophilin A (TCyp), are attractive candidates owing to their potency in sensing HIV-1 and blocking its activity. Here, we show that expression of RhT5 and TCyp in HIV-1-infected cells improves CD8(+) T-cell-mediated inhibition through the direct activation of HIV-1-specific CD8(+) T-cell responses. We found that the potency in CD8(+) activation was stronger for RhT5 variants and capsid-specific CD8(+) T cells in a mechanism that relies on TRIM5-dependent particle recruitment to cellular proteasomes. This novel mechanism couples innate viral sensing with cellular immunity in a single protein and could be exploited to develop innovative therapeutics for control of HIV-1 infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cyclophilin A/metabolism , HIV-1/immunology , Macaca mulatta/immunology , Proteins/metabolism , Animals , Cell Line , Humans , Ubiquitin-Protein Ligases
9.
J Neurosci ; 35(32): 11221-32, 2015 Aug 12.
Article in English | MEDLINE | ID: mdl-26269632

ABSTRACT

Spiral ganglion neurons (SGNs) relay acoustic code from cochlear hair cells to the brainstem, and their stimulation enables electrical hearing via cochlear implants. Rapid adaptation, a mechanism that preserves temporal precision, and a prominent feature of auditory neurons, is regulated via dendrotoxin-sensitive low-threshold voltage-activated (LVA) K(+) channels. Here, we investigated the molecular physiology of LVA currents in SGNs cultured from mice following the onset of hearing (postnatal days 12-21). Kv1.1- and Kv1.2-specific toxins blocked the LVA currents in a comparable manner, suggesting that both subunits contribute to functional heteromeric channels. Confocal immunofluorescence in fixed cochlear sections localized both Kv1.1 and Kv1.2 subunits to specific neuronal microdomains, including the somatic membrane, juxtaparanodes, and the first heminode, which forms the spike initiation site of the auditory nerve. The spatial distribution of Kv1 immunofluorescence appeared mutually exclusive to that of Kv3.1b subunits, which mediate high-threshold voltage-activated currents. As Kv1.2-containing channels are positively modulated by membrane phosphoinositides, we investigated the influence of phosphatidylinositol-4,5-bisphosphate (PIP2) availability on SGN electrophysiology. Reducing PIP2 production using wortmannin, or sequestration of PIP2 using a palmitoylated peptide (PIP2-PP), slowed adaptation rate in SGN populations. PIP2-PP specifically inhibited the LVA current in SGNs, an effect reduced by intracellular dialysis of a nonhydrolysable analog of PIP2. PIP2-PP also inhibited heterologously expressed Kv1.1/Kv1.2 channels, recapitulating its effect in SGNs. Collectively, the data identify Kv1.1/Kv1.2 heteromeric channels as key regulators of action potential initiation and propagation in the auditory nerve, and suggest that modulation of these channels by endogenous phosphoinositides provides local control of membrane excitability. SIGNIFICANCE STATEMENT: Rapid spike adaptation is an important feature of auditory neurons that preserves temporal precision. In spiral ganglion neurons, the primary afferents in the cochlea, adaptation is regulated by heteromeric ion channels composed of Kv1.1 and Kv1.2 subunits. These subunits colocalize to common functional microdomains, such as juxtaparanodes and the somatic membrane. Activity of the heteromeric channels is controlled by cellular availability of PIP2, a membrane phospholipid. This mechanism provides an intrinsic regulation of output from the auditory nerve, which could be targeted for therapeutic adjustment of hearing sensitivity.


Subject(s)
Action Potentials/physiology , Kv1.1 Potassium Channel/metabolism , Kv1.2 Potassium Channel/metabolism , Neurons/physiology , Phosphatidylinositol 4,5-Diphosphate/metabolism , Spiral Ganglion/physiology , Action Potentials/drug effects , Androstadienes/pharmacology , Animals , Female , Hearing/physiology , Male , Mice , Neurons/drug effects , Neurons/metabolism , Spiral Ganglion/drug effects , Spiral Ganglion/metabolism , Wortmannin
10.
FASEB J ; 28(1): 117-30, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24121462

ABSTRACT

The purpose of this study was the generation of central nervous system (CNS)-excluded cannabinoid receptor agonists to test the hypothesis that inhibition of spasticity, due to CNS autoimmunity, could be controlled by affecting neurotransmission within the periphery. Procedures included identification of chemicals and modeling to predict the mode of exclusion; induction and control of spasticity in the ABH mouse model of multiple sclerosis; conditional deletion of CB1 receptor in peripheral nerves; side-effect profiling to demonstrate the mechanism of CNS-exclusion via drug pumps; genome-wide association study in N2(129×ABH) backcross to map polymorphic cannabinoid drug pump; and sequencing and detection of cannabinoid drug-pump activity in human brain endothelial cell lines. Three drugs (CT3, SAB378 and SAD448) were identified that control spasticity via action on the peripheral nerve CB1 receptor. These were peripherally restricted via drug pumps that limit the CNS side effects (hypothermia) of cannabinoids to increase the therapeutic window. A cannabinoid drug pump is polymorphic and functionally lacking in many laboratory (C57BL/6, 129, CD-1) mice used for transgenesis, pharmacology, and toxicology studies. This phenotype was mapped and controlled by 1-3 genetic loci. ABCC1 within a cluster showing linkage is a cannabinoid CNS-drug pump. Global and conditional CB1 receptor-knockout mice were used as controls. In summary, CNS-excluded CB1 receptor agonists are a novel class of therapeutic agent for spasticity.


Subject(s)
Cannabinoid Receptor Agonists/pharmacology , Central Nervous System/drug effects , Multiple Sclerosis/drug therapy , Muscle Spasticity/drug therapy , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/metabolism , Animals , Cannabinoids/metabolism , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Female , Mice , Multidrug Resistance-Associated Proteins/metabolism
11.
Bioorg Med Chem ; 23(17): 5303-10, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26264842

ABSTRACT

Soluble guanylate cyclase (sGC) is a haem containing enzyme that regulates cardiovascular homeostasis and multiple mechanisms in the central and peripheral nervous system. Commonly used inhibitors of sGC activity act through oxidation of the haem moiety, however they also bind haemoglobin and this limits their bioavailability for in vivo studies. We have discovered a new class of small molecule inhibitors of sGC and have characterised a compound designated D12 (compound 10) which binds to the catalytic domain of the enzyme with a KD of 11 µM in a SPR assay.


Subject(s)
Enzyme Activators/chemistry , Enzyme Activators/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Quinoxalines/chemistry , Quinoxalines/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Catalytic Domain , Guanylate Cyclase/chemistry , Guanylate Cyclase/metabolism , Humans , Molecular Docking Simulation , Nitric Oxide/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Soluble Guanylyl Cyclase
12.
Brain ; 137(Pt 1): 92-108, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24287115

ABSTRACT

Progressive multiple sclerosis is associated with metabolic failure of the axon and excitotoxicity that leads to chronic neurodegeneration. Global sodium-channel blockade causes side effects that can limit its use for neuroprotection in multiple sclerosis. Through selective targeting of drugs to lesions we aimed to improve the potential therapeutic window for treatment. This was assessed in the relapsing-progressive experimental autoimmune encephalomyelitis ABH mouse model of multiple sclerosis using conventional sodium channel blockers and a novel central nervous system-excluded sodium channel blocker (CFM6104) that was synthesized with properties that selectively target the inflammatory penumbra in experimental autoimmune encephalomyelitis lesions. Carbamazepine and oxcarbazepine were not immunosuppressive in lymphocyte-driven autoimmunity, but slowed the accumulation of disability in experimental autoimmune encephalomyelitis when administered during periods of the inflammatory penumbra after active lesion formation, and was shown to limit the development of neurodegeneration during optic neuritis in myelin-specific T cell receptor transgenic mice. CFM6104 was shown to be a state-selective, sodium channel blocker and a fluorescent p-glycoprotein substrate that was traceable. This compound was >90% excluded from the central nervous system in normal mice, but entered the central nervous system during the inflammatory phase in experimental autoimmune encephalomyelitis mice. This occurs after the focal and selective downregulation of endothelial p-glycoprotein at the blood-brain barrier that occurs in both experimental autoimmune encephalomyelitis and multiple sclerosis lesions. CFM6104 significantly slowed down the accumulation of disability and nerve loss in experimental autoimmune encephalomyelitis. Therapeutic-targeting of drugs to lesions may reduce the potential side effect profile of neuroprotective agents that can influence neurotransmission. This class of agents inhibit microglial activity and neural sodium loading, which are both thought to contribute to progressive neurodegeneration in multiple sclerosis and possibly other neurodegenerative diseases.


Subject(s)
Benzamides/therapeutic use , Indazoles/therapeutic use , Multiple Sclerosis/drug therapy , Neuroprotective Agents/therapeutic use , Oxadiazoles/therapeutic use , Sodium Channel Blockers/therapeutic use , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Biological Specimen Banks , Brain/pathology , Carbamazepine/pharmacology , Carrier Proteins/metabolism , Cell Proliferation/drug effects , Chromatography, High Pressure Liquid , Drug Delivery Systems , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Immunohistochemistry , Mass Spectrometry , Mice , Mice, Inbred C57BL , Multiple Sclerosis/physiopathology , Optic Neuritis/physiopathology , T-Lymphocytes/drug effects , Uveitis/physiopathology , Voltage-Gated Sodium Channels/metabolism
13.
Chembiochem ; 15(8): 1161-70, 2014 May 26.
Article in English | MEDLINE | ID: mdl-24771685

ABSTRACT

The interaction between VEGF-A and its neuropilin (NRP) receptors mediates a number of important biological effects. NRP1 and the related molecule NRP2 are widely expressed on multiple tumour types and throughout the tumour vasculature, and are emerging as critical molecules required for the progression of angiogenic diseases. Given the increasing evidence supporting a role for NRP1 in tumour development, there is growing interest in developing inhibitors of NRP1 interactions with VEGF and its other ligands. In order to probe the interaction we synthesised a number of exon 7- and 8-derived bicyclic peptides with N-terminal lipophilic groups and found a simple N-octanoyl derivative (EG00086) to be the most potent and functionally active. Detailed modelling studies indicated that new intramolecular hydrogen bonds were formed, stabilising the structure and possibly contributing to the potency. Removal of a salt bridge between D142 and R164 implicated in VEGF-A binding to neuropilin-1 had a minor effect on potency. Isothermal calorimetry was used to assess binding of EG00086 to NRP1 and NRP2, and the stability of the peptide in serum and in vivo was investigated. EG00086 is a potent blocker of VEGF-promoted cellular adhesion to extracellular matrices, and phosphorylation of p130Cas contributes to this effect.


Subject(s)
Neuropilin-1/metabolism , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/metabolism , Binding Sites , Cell Adhesion/drug effects , Cell Survival/drug effects , Cells, Cultured , Crk-Associated Substrate Protein/metabolism , Exons/genetics , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Humans , Lipopeptides/chemistry , Lipopeptides/metabolism , Lipopeptides/pharmacology , Molecular Dynamics Simulation , Neuropilin-1/chemistry , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/pharmacology , Phosphorylation/drug effects , Protein Binding , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics
14.
J Antimicrob Chemother ; 69(4): 1017-26, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24324225

ABSTRACT

OBJECTIVES: To identify and to characterize small-molecule inhibitors that target the subunit polymerization of the type 1 pilus assembly in uropathogenic Escherichia coli (UPEC). METHODS: Using an SDS-PAGE-based assay, in silico pre-filtered small-molecule compounds were screened for specific inhibitory activity against the critical subunit polymerization step of the chaperone-usher pathway during pilus biogenesis. The biological activity of one of the compounds was validated in assays monitoring UPEC type 1 pilus biogenesis, type 1 pilus-dependent biofilm formation and adherence to human bladder epithelial cells. The time dependence of the in vivo inhibitory activity and the overall effect of the compound on UPEC growth were determined. RESULTS: N-(4-chloro-phenyl)-2-{5-[4-(pyrrolidine-1-sulfonyl)-phenyl]-[1,3,4]oxadiazol-2-yl sulfanyl}-acetamide (AL1) inhibited in vitro pilus subunit polymerization. In bacterial cultures, AL1 disrupted UPEC type 1 pilus biogenesis and pilus-dependent biofilm formation, and resulted in the reduction of bacterial adherence to human bladder epithelial cells, without affecting bacterial cell growth. Bacterial exposure to the inhibitor led to an almost instantaneous loss of type 1 pili. CONCLUSIONS: We have identified and characterized a small molecule that interferes with the assembly of type 1 pili. The molecule targets the polymerization step during the subunit incorporation cycle of the chaperone-usher pathway. Our discovery provides new insight into the design and development of novel anti-virulence therapies targeting key virulence factors of bacterial pathogens.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Adhesion/drug effects , Fimbriae, Bacterial/drug effects , Macromolecular Substances/metabolism , Protein Multimerization/drug effects , Protein Subunits/metabolism , Uropathogenic Escherichia coli/drug effects , Animals , Biofilms/drug effects , Cell Line , Epithelial Cells/microbiology , Humans , Uropathogenic Escherichia coli/physiology
15.
Bioorg Med Chem Lett ; 24(4): 1075-9, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24480469

ABSTRACT

Soluble Guanylate Cyclase (sGC) is the receptor for the signalling agent nitric oxide (NO) and catalyses the production of the second messenger cyclic guanosine monophosphate (cGMP) from guanosine triphosphate (GTP). The enzyme is an attractive drug target for small molecules that act in the cardiovascular and pulmonary systems, and has also shown to be a potential target in neurological disorders. We have discovered that 5-(indazol-3-yl)-1,2,4-oxadiazoles activate the enzyme in the absence of added NO and shown they bind to the catalytic domain of the enzyme after development of a surface plasmon resonance assay that allows the biophysical detection of intrinsic binding of ligands to the full length sGC and to a construct of the catalytic domain.


Subject(s)
Guanylate Cyclase/metabolism , Oxadiazoles/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Surface Plasmon Resonance , Biocatalysis , Catalytic Domain/drug effects , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Guanosine Monophosphate/biosynthesis , Guanylate Cyclase/antagonists & inhibitors , Molecular Structure , Oxadiazoles/chemistry , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Soluble Guanylyl Cyclase , Structure-Activity Relationship
16.
Bioorg Med Chem ; 22(22): 6459-70, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25438770

ABSTRACT

A novel series of 8-amino imidazo[1,2-a]pyrazine derivatives has been developed as inhibitors of the VirB11 ATPase HP0525, a key component of the bacterial type IV secretion system. A flexible synthetic route to both 2- and 3-aryl substituted regioisomers has been developed. The resulting series of imidazo[1,2-a]pyrazines has been used to probe the structure-activity relationships of these inhibitors, which show potential as antibacterial agents.


Subject(s)
Anti-Bacterial Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Imidazoles/chemistry , Pyrazines/chemistry , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/metabolism , Bacterial Proteins/metabolism , Binding Sites , Gram-Negative Bacteria/metabolism , Imidazoles/chemical synthesis , Imidazoles/metabolism , Kinetics , Molecular Docking Simulation , Protein Binding , Protein Structure, Tertiary , Pyrazines/chemical synthesis , Pyrazines/metabolism , Structure-Activity Relationship
17.
Sci Adv ; 10(28): eado3501, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38985859

ABSTRACT

Macrocyclic drugs can address an increasing range of molecular targets but enabling central nervous system (CNS) access to these drugs has been viewed as an intractable problem. We designed and synthesized a series of quinolinium-modified cyclosporine derivatives targeted to the mitochondrial cyclophilin D protein. Modification of the cation to enable greater delocalization was confirmed by x-ray crystallography of the cations. Critically, greater delocalization improved brain concentrations. Assessment of the compounds in preclinical assays and for pharmacokinetics identified a molecule JP1-138 with at least 20 times the brain levels of a non-delocalized compound or those reported for cyclosporine. Levels were maintained over 24 hours together with low hERG potential. The paradigm outlined here could have widespread utility in the treatment of CNS diseases.


Subject(s)
Quinolinium Compounds , Animals , Humans , Quinolinium Compounds/chemistry , Quinolinium Compounds/pharmacokinetics , Cyclosporine/chemistry , Cyclosporine/pharmacokinetics , Central Nervous System/metabolism , Central Nervous System/drug effects , Crystallography, X-Ray , Peptides/chemistry , Peptides/pharmacokinetics , Brain/metabolism , Brain/drug effects , Mice
18.
J Neurochem ; 127(3): 394-402, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24033337

ABSTRACT

Tuftsin (Thr-Lys-Pro-Arg) is a natural immunomodulating peptide found to stimulate phagocytosis in macrophages/microglia. Tuftsin binds to the receptor neuropilin-1 (Nrp1) on the surface of cells. Nrp1 is a single-pass transmembrane protein, but its intracellular C-terminal domain is too small to signal independently. Instead, it associates with a variety of coreceptors. Despite its long history, the pathway through which tuftsin signals has not been described. To investigate this question, we employed various inhibitors to Nrp1's coreceptors to determine which route is responsible for tuftsin signaling. We use the inhibitor EG00229, which prevents tuftsin binding to Nrp1 on the surface of microglia and reverses the anti-inflammatory M2 shift induced by tuftsin. Furthermore, we demonstrate that blockade of transforming growth factor beta (TGFß) signaling via TßR1 disrupts the M2 shift similar to EG00229. We report that tuftsin promotes Smad3 phosphorylation and reduces Akt phosphorylation. Taken together, our data show that tuftsin signals through Nrp1 and the canonical TGFß signaling pathway. Despite the 40-year history of the tetrapeptide tuftsin (TKPR), a macrophage and microglial activator, its mechanism of action has not been defined. Here, we report that the tuftsin-mediated anti-inflammatory M2 shift in microglia is caused specifically by tuftsin binding to the receptor neuropilin-1 (Nrp1) and signaling through TGFß receptor-1, a coreceptor of Nrp1. We further show that tuftsin signals via the canonical TGFß pathway and promotes TGFß release from target cells.


Subject(s)
Immunologic Factors/physiology , Neuropilin-1/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Tuftsin/physiology , Animals , Blotting, Western , Cerebral Cortex/cytology , Cytokines/metabolism , Fluorescent Antibody Technique , Immunologic Factors/metabolism , Methionine/physiology , Mice , Mice, Inbred C57BL , Microglia/metabolism , Neurons/physiology , Neuropilin-1/antagonists & inhibitors , Nitric Oxide/metabolism , Primary Cell Culture , RNA/biosynthesis , RNA/isolation & purification , Real-Time Polymerase Chain Reaction , Receptors, Cell Surface/metabolism , Tuftsin/metabolism
19.
Pharmaceuticals (Basel) ; 16(7)2023 Jul 07.
Article in English | MEDLINE | ID: mdl-37513884

ABSTRACT

Big conductance calcium-activated (BK) channel openers can inhibit pathologically driven neural hyperactivity to control symptoms via hyperpolarizing signals to limit neural excitability. We hypothesized that BK channel openers would be neuroprotective during neuroinflammatory, autoimmune disease. The neurodegenerative disease was induced in a mouse experimental autoimmune encephalomyelitis model with translational value to detect neuroprotection in multiple sclerosis. Following the treatment with the BK channel openers, BMS-204253 and VSN16R, neuroprotection was assessed using subjective and objective clinical outcomes and by quantitating spinal nerve content. Treatment with BMS-204253 and VSN16R did not inhibit the development of relapsing autoimmunity, consistent with minimal channel expression via immune cells, nor did it change leukocyte levels in rodents or humans. However, it inhibited the accumulation of nerve loss and disability as a consequence of autoimmunity. Therefore, in addition to symptom control, BK channel openers have the potential to save nerves from excitotoxic damage and could be useful as either stand-alone neuroprotective agents or as add-ons to current disease-modifying treatments that block relapsing MS but do not have any direct neuroprotective activity.

20.
Eur J Cell Biol ; 101(1): 151185, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34915361

ABSTRACT

The PINK1/Parkin pathway plays an important role in maintaining a healthy pool of mitochondria. Activation of this pathway can lead to apoptosis, mitophagy, or mitochondrial-derived vesicle formation, depending on the nature of mitochondrial damage. The signaling by which PINK/Parkin activation leads to these different mitochondrial outcomes remains understudied. Here we present evidence that cannabidiol (CBD) activates the PINK1-Parkin pathway in a unique manner. CBD stimulates PINK1-dependent Parkin mitochondrial recruitment similarly to other well-studied Parkin activators but with a distinctive shift in the temporal dynamics and mitochondrial fates. The mitochondrial permeability transition pore inhibitor cyclosporine A exclusively diminished the CBD-induced PINK1/Parkin activation and its associated mitochondrial effects. Unexpectedly, CBD treatment also induced elevated production of mitochondrial-derived vesicles (MDV), a potential quality control mechanism that may help repair partial damaged mitochondria. Our results suggest that CBD may engage the PINK1-Parkin pathway to produce MDV and repair mitochondrial lesions via mitochondrial permeability transition pore opening. This work uncovered a novel link between CBD and PINK1/Parkin-dependent MDV production in mitochondrial health regulation.


Subject(s)
Cannabidiol , Mitophagy , Mitochondria , Protein Kinases , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL