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










Publication year range
1.
Front Immunol ; 13: 956603, 2022.
Article in English | MEDLINE | ID: mdl-36389776

ABSTRACT

Tapasin, a component of the major histocompatibility complex (MHC) I peptide loading complex, edits the repertoire of peptides that is presented at the cell surface by MHC I and thereby plays a key role in shaping the hierarchy of CD8+ T-cell responses to tumors and pathogens. We have developed a system that allows us to tune the level of tapasin expression and independently regulate the expression of competing peptides of different off-rates. By quantifying the relative surface expression of peptides presented by MHC I molecules, we show that peptide editing by tapasin can be measured in terms of "tapasin bonus," which is dependent on both peptide kinetic stability (off-rate) and peptide abundance (peptide supply). Each peptide has therefore an individual tapasin bonus fingerprint. We also show that there is an optimal level of tapasin expression for each peptide in the immunopeptidome, dependent on its off-rate and abundance. This is important, as the level of tapasin expression can vary widely during different stages of the immune response against pathogens or cancer and is often the target for immune escape.


Subject(s)
Histocompatibility Antigens Class I , Peptides , Epitopes , Histocompatibility Antigens , Major Histocompatibility Complex
2.
ACS Pharmacol Transl Sci ; 5(11): 1169-1180, 2022 Nov 11.
Article in English | MEDLINE | ID: mdl-36407959

ABSTRACT

The tumor suppressor protein p53 is inactivated in the majority of human cancers and remains a prime target for developing new drugs to reactivate its tumor suppressing activity for anticancer therapies. The oncogenic p53 mutant Y220C accounts for approximately 125,000 new cancer cases per annum and is one of the most prevalent p53 mutants overall. It harbors a narrow, mutationally induced pocket at the surface of the DNA-binding domain that destabilizes p53, leading to its rapid denaturation and aggregation. Here, we present the structure-guided development of high-affinity small molecules stabilizing p53-Y220C in vitro, along with the synthetic routes developed in the process, in vitro structure-activity relationship data, and confirmation of their binding mode by protein X-ray crystallography. We disclose two new chemical probes displaying sub-micromolar binding affinity in vitro, marking an important milestone since the discovery of the first small-molecule ligand of Y220C in 2008. New chemical probe JC744 displayed a K d = 320 nM, along with potent in vitro protein stabilization. This study, therefore, represents a significant advance toward high-affinity Y220C ligands for clinical evaluation.

3.
Commun Biol ; 4(1): 772, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162985

ABSTRACT

Monoclonal antibodies (mAb) and natural ligands targeting costimulatory tumor necrosis factor receptors (TNFR) exhibit a wide range of agonistic activities and antitumor responses. The mechanisms underlying these differential agonistic activities remain poorly understood. Here, we employ a panel of experimental and clinically-relevant molecules targeting human CD40, 4-1BB and OX40 to examine this issue. Confocal and STORM microscopy reveal that strongly agonistic reagents induce clusters characterized by small area and high receptor density. Using antibody pairs differing only in isotype we show that hIgG2 confers significantly more receptor clustering than hIgG1 across all three receptors, explaining its greater agonistic activity, with receptor clustering shielding the receptor-agonist complex from further molecular access. Nevertheless, discrete receptor clustering patterns are observed with different hIgG2 mAb, with a unique rod-shaped assembly observed with the most agonistic mAb. These findings dispel the notion that larger receptor clusters elicit greater agonism, and instead point to receptor density and subsequent super-structure as key determinants.


Subject(s)
Receptors, Tumor Necrosis Factor/agonists , Animals , Antibodies, Monoclonal/pharmacology , Antibody Affinity , CD40 Antigens/agonists , CD40 Antigens/chemistry , Cell Line , Humans , Immunoglobulin G/pharmacology , Mice , Microscopy, Confocal , Receptors, OX40/agonists , Receptors, Tumor Necrosis Factor/chemistry , Tumor Necrosis Factor Receptor Superfamily, Member 9/agonists
4.
ChemMedChem ; 16(8): 1316-1324, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33533576

ABSTRACT

Lysine-specific demethylase 1 (LSD1/KDM1A) oxidatively removes methyl groups from histone proteins, and its aberrant activity has been correlated with cancers including acute myeloid leukemia (AML). We report a novel series of tranylcypromine analogues with a carboxamide at the 4-position of the aryl ring. These compounds, such as 5 a and 5 b with benzyl and phenethylamide substituents, respectively, had potent sub-micromolar IC50 values for the inhibition of LSD1 as well as cell proliferation in a panel of AML cell lines. The dose-dependent increase in cellular expression levels of H3K4me2, CD86, CD11b and CD14 supported a mechanism involving LSD1 inhibition. The tert-butyl and ethyl carbamate derivatives of these tranylcypromines, although inactive in LSD1 inhibition, were of similar potency in cell-based assays with a more rapid onset of action. This suggests that carbamates can act as metabolically labile tranylcypromine prodrugs with superior pharmacokinetics.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Tranylcypromine/analogs & derivatives , Tranylcypromine/pharmacology , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Humans
5.
Cancer Cell ; 37(6): 850-866.e7, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32442402

ABSTRACT

Anti-CD40 monoclonal antibodies (mAbs) comprise agonists and antagonists, which display promising therapeutic activities in cancer and autoimmunity, respectively. We previously showed that epitope and isotype interact to deliver optimal agonistic anti-CD40 mAbs. The impact of Fc engineering on antagonists, however, remains largely unexplored. Here, we show that clinically relevant antagonists used for treating autoimmune conditions can be converted into potent FcγR-independent agonists with remarkable antitumor activity by isotype switching to hIgG2. One antagonist is converted to a super-agonist with greater potency than previously reported highly agonistic anti-CD40 mAbs. Such conversion is dependent on the unique disulfide bonding properties of the hIgG2 hinge. This investigation highlights the transformative capacity of the hIgG2 isotype for converting antagonists to agonists to treat cancer.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD40 Antigens/immunology , CD40 Ligand/immunology , Dendritic Cells/immunology , Immunoglobulin Class Switching/immunology , Immunoglobulin G/immunology , Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/immunology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Dendritic Cells/drug effects , Immunoglobulin Class Switching/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Receptors, IgE/physiology , Receptors, IgG/physiology , Thymus Neoplasms/drug therapy , Thymus Neoplasms/immunology , Thymus Neoplasms/metabolism , Thymus Neoplasms/pathology
6.
Cells ; 8(3)2019 03 02.
Article in English | MEDLINE | ID: mdl-30832318

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is a common and deadly cancer; however, very little improvement has been made towards its diagnosis and prognosis. The expression and functional contribution of the receptor tyrosine kinase ROR1 have not been investigated in HCC before. Hence, we investigated the expression of ROR1 in HCC cells and assessed its involvement in hepatocarcinogenesis. METHODS: Recombinant bacterial ROR1 protein was used as an immunogen to generate ROR1 monoclonal antibodies. ROR1 transcript levels were detected by RT-qPCR and the protein expression of ROR1 in HCC was assessed by Western blotting by using homemade anti-ROR1 monoclonal antibodies. Apoptosis, cell cycle, trans-well migration, and drug efflux assays were performed in shRNA-ROR1 HCC cell clones to uncover the functional contribution of ROR1 to hepatocarcinogenesis. RESULTS: New ROR1 antibodies specifically detected endogenous ROR1 protein in human and mouse HCC cell lines. ROR1-knockdown resulted in decreased proliferation and migration but enhanced resistance to apoptosis and anoikis. The observed chemotherapy-resistant phenotype of ROR1-knockdown cells was due to enhanced drug efflux and increased expression of multi-drug resistance genes. CONCLUSIONS: ROR1 is expressed in HCC and contributes to disease development by interfering with multiple pathways. Acquired ROR1 expression may have diagnostic and prognostic value in HCC.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Liver Neoplasms/enzymology , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Animals , Anoikis/drug effects , Anoikis/genetics , Antibodies, Monoclonal/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/drug effects , G1 Phase/drug effects , G1 Phase/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Phenotype , Up-Regulation/drug effects , Up-Regulation/genetics
7.
Cancer Cell Int ; 18: 71, 2018.
Article in English | MEDLINE | ID: mdl-29760584

ABSTRACT

BACKGROUND: Castrate resistant prostate cancer (CRPC) is often driven by constitutively active forms of the androgen receptor such as the V7 splice variant (AR-V7) and commonly becomes resistant to established hormonal therapy strategies such as enzalutamide as a result. The lysine demethylase LSD1 is a co-activator of the wild type androgen receptor and a potential therapeutic target in hormone sensitive prostate cancer. We evaluated whether LSD1 could also be therapeutically targeted in CRPC models driven by AR-V7. METHODS: We utilised cell line models of castrate resistant prostate cancer through over expression of AR-V7 to test the impact of chemical LSD1 inhibition on AR activation. We validated findings through depletion of LSD1 expression and in prostate cancer cell lines that express AR-V7. RESULTS: Chemical inhibition of LSD1 resulted in reduced activation of the androgen receptor through both the wild type and its AR-V7 splice variant forms. This was confirmed and validated in luciferase reporter assays, in LNCaP and 22Rv1 prostate cancer cell lines and in LSD1 depletion experiments. CONCLUSION: LSD1 contributes to activation of both the wild type and V7 splice variant forms of the androgen receptor and can be therapeutically targeted in models of CRPC. Further development of this approach is warranted.

8.
Cancer Cell ; 33(4): 664-675.e4, 2018 04 09.
Article in English | MEDLINE | ID: mdl-29576376

ABSTRACT

Anti-CD40 monoclonal antibodies (mAbs) that promote or inhibit receptor function hold promise as therapeutics for cancer and autoimmunity. Rules governing their diverse range of functions, however, are lacking. Here we determined characteristics of nine hCD40 mAbs engaging epitopes throughout the CD40 extracellular region expressed as varying isotypes. All mAb formats were strong agonists when hyper-crosslinked; however, only those binding the membrane-distal cysteine-rich domain 1 (CRD1) retained agonistic activity with physiological Fc gamma receptor crosslinking or as human immunoglobulin G2 isotype; agonistic activity decreased as epitopes drew closer to the membrane. In addition, all CRD2-4 binding mAbs blocked CD40 ligand interaction and were potent antagonists. Thus, the membrane distal CRD1 provides a region of choice for selecting CD40 agonists while CRD2-4 provides antagonistic epitopes.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD40 Antigens/chemistry , CD40 Antigens/metabolism , Epitopes/chemistry , Antibodies, Monoclonal/chemistry , Antibody Specificity , CD40 Antigens/agonists , CD40 Ligand/metabolism , Cross-Linking Reagents , Humans , Models, Molecular , Protein Binding/drug effects
9.
J Biol Chem ; 293(20): 7538-7548, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29599287

ABSTRACT

Endoplasmic reticulum aminopeptidase 1 (ERAP1) and ERAP2 process N-terminally extended antigenic precursors for optimal loading onto major histocompatibility complex class I (MHC I) molecules. We and others have demonstrated that ERAP1 processes peptides bound to MHC I, but the underlying mechanism is unknown. To this end, we utilized single-chain trimers (SCT) of the ovalbumin-derived epitope SIINFEKL (SL8) tethered to the H2-Kb MHC I determinant from mouse and introduced three substitutions, E63A, K66A, and W167A, at the A-pocket of the peptide-binding groove in the MHC I heavy chain, which interact with the N termini of peptides. These variants significantly decreased SL8-presenting SCT at the cell surface in the presence of ERAP1, but did not affect overall SCT expression, indicating that ERAP1 trims the SL8 N terminus. Comparison of the X-ray crystal structures of WT and three variant SCTs revealed only minor perturbations of the peptide-binding domain in the variants. However, molecular dynamics simulations suggested that SL8 can dissociate partially within a sub-microsecond timescale, exposing its N terminus to the solvent. We also found that the C terminus of MHC I-bound SL8 remains deeply buried in the F-pocket of MHC I. Furthermore, free-energy calculations revealed that the three SCT variants exhibit lower free-energy barriers of N terminus dissociation than the WT Kb Taken together, our results are consistent with a previously observed model in which the partial dissociation of bound peptides from MHC I exposes their N terminus to trimming by ERAP1, whereas their C terminus is anchored at the F-pocket.


Subject(s)
Aminopeptidases/metabolism , Epitopes/metabolism , Histocompatibility Antigens Class I/metabolism , Minor Histocompatibility Antigens/metabolism , Aminopeptidases/chemistry , Antigen Presentation , Crystallography, X-Ray , Epitopes/chemistry , HeLa Cells , Histocompatibility Antigens Class I/chemistry , Humans , Minor Histocompatibility Antigens/chemistry , Models, Molecular , Protein Conformation , Protein Domains
10.
Cancer Immunol Immunother ; 67(4): 627-638, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29330557

ABSTRACT

Vaccination with DNA that encodes cancer antigens is a simple and convenient way to raise immunity against cancer and has already shown promise in the clinical setting. Conventional plasmid DNA is commonly used which together with the encoded antigen also includes bacterial immunostimulatory CpG motifs to target the DNA sensor Toll-like receptor 9. Recently DNA vaccines using doggybone DNA (dbDNA™), have been developed without the use of bacteria. The cell-free process relies on the use of Phi29 DNA polymerase to amplify the template followed by protelomerase TelN to complete individual closed linear DNA. The resulting DNA contains the required antigenic sequence, a promoter and a poly A tail but lacks bacterial sequences such as an antibiotic resistance gene, prompting the question of immunogenicity. Here we compared the ability of doggybone DNA vaccine with plasmid DNA vaccine to induce adaptive immunity using clinically relevant oncotargets E6 and E7 from HPV. We demonstrate that despite the inability to trigger TLR9, doggybone DNA was able to induce similar levels of cellular and humoral immunity as plasmid DNA, with suppression of established TC-1 tumours.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immunity, Cellular/immunology , Lung Neoplasms/immunology , Plasmids/immunology , Toll-Like Receptor 9/immunology , Vaccines, DNA/immunology , Animals , Disease Models, Animal , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Mice , Mice, Inbred C57BL , Plasmids/administration & dosage , Plasmids/genetics , Tumor Cells, Cultured , Vaccination , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
11.
Bioorg Med Chem Lett ; 27(10): 2099-2101, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28390942

ABSTRACT

We report a series of tranylcypromine analogues containing a fluorine in the cyclopropyl ring. A number of compounds with additional m- or p-substitution of the aryl ring were micromolar inhibitors of the LSD1 enzyme. In cellular assays, the compounds inhibited the proliferation of acute myeloid leukemia cell lines. Increased levels of the biomarkers H3K4me2 and CD86 were consistent with LSD1 target engagement.


Subject(s)
Enzyme Inhibitors/chemistry , Histone Demethylases/antagonists & inhibitors , Tranylcypromine/analogs & derivatives , B7-2 Antigen/metabolism , Biomarkers/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/toxicity , Halogenation , Histone Demethylases/metabolism , Histones/metabolism , Humans , Inhibitory Concentration 50 , Tranylcypromine/chemical synthesis , Tranylcypromine/toxicity
12.
Elife ; 52016 10 26.
Article in English | MEDLINE | ID: mdl-27782881

ABSTRACT

The auxiliary α2δ subunits of voltage-gated calcium channels are extracellular membrane-associated proteins, which are post-translationally cleaved into disulfide-linked polypeptides α2 and δ. We now show, using α2δ constructs containing artificial cleavage sites, that this processing is an essential step permitting voltage-dependent activation of plasma membrane N-type (CaV2.2) calcium channels. Indeed, uncleaved α2δ inhibits native calcium currents in mammalian neurons. By inducing acute cell-surface proteolytic cleavage of α2δ, voltage-dependent activation of channels is promoted, independent from the trafficking role of α2δ. Uncleaved α2δ does not support trafficking of CaV2.2 channel complexes into neuronal processes, and inhibits Ca2+ entry into synaptic boutons, and we can reverse this by controlled intracellular proteolytic cleavage. We propose a model whereby uncleaved α2δ subunits maintain immature calcium channels in an inhibited state. Proteolytic processing of α2δ then permits voltage-dependent activation of the channels, acting as a checkpoint allowing trafficking only of mature calcium channel complexes into neuronal processes.


Subject(s)
Calcium Channels, N-Type/metabolism , Neurons/enzymology , Protein Processing, Post-Translational , Animals , Mice , Models, Biological , Protein Transport , Proteolysis , Rabbits , Rats
13.
Sci Rep ; 6: 22237, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26928456

ABSTRACT

Google Glass is a recently designed wearable device capable of displaying information in a smartphone-like hands-free format by wireless communication. The Glass also provides convenient control over remote devices, primarily enabled by voice recognition commands. These unique features of the Google Glass make it useful for medical and biomedical applications where hands-free experiences are strongly preferred. Here, we report for the first time, an integral set of hardware, firmware, software, and Glassware that enabled wireless transmission of sensor data onto the Google Glass for on-demand data visualization and real-time analysis. Additionally, the platform allowed the user to control outputs entered through the Glass, therefore achieving bi-directional Glass-device interfacing. Using this versatile platform, we demonstrated its capability in monitoring physical and physiological parameters such as temperature, pH, and morphology of liver- and heart-on-chips. Furthermore, we showed the capability to remotely introduce pharmaceutical compounds into a microfluidic human primary liver bioreactor at desired time points while monitoring their effects through the Glass. We believe that such an innovative platform, along with its concept, has set up a premise in wearable monitoring and controlling technology for a wide variety of applications in biomedicine.


Subject(s)
Lab-On-A-Chip Devices/statistics & numerical data , Monitoring, Physiologic/methods , Speech Recognition Software , Telemedicine , Actuarial Analysis , Biosensing Techniques , Humans , Microfluidic Analytical Techniques , Quality Control , Smartphone , Telemedicine/trends , User-Computer Interface , Wireless Technology
14.
Cancer Immunol Immunother ; 62(6): 1093-105, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23604105

ABSTRACT

Attempts to generate robust anti-tumour cytotoxic T lymphocyte (CTL) responses using immunotherapy are frequently thwarted by exhaustion and anergy of CTL recruited to tumour. One strategy to overcome this is to retarget a population of virus-specific CTL to kill tumour cells. Here, we describe a proof-of-principle study using a bispecific conjugate designed to retarget ovalbumin (OVA)-specific CTL to kill tumour cells via CD20. A single-chain trimer (SCT) consisting of MHCI H-2K(b)/SIINFEKL peptide/beta 2 microglobulin/BirA was expressed in bacteria, refolded and chemically conjugated to one (1:1; F2) or two (2:1; F3) anti-hCD20 Fab' fragments. In vitro, the [SCT × Fab'] (F2 and F3) redirected SIINFEKL-specific OT-I CTL to kill CD20(+) target cells, and in the presence of CD20(+) target cells to provide crosslinking, they were also able to induce proliferation of OT-I cells. In vivo, activated OT-I CTL could be retargeted to kill [SCT × Fab']-coated B cells from hCD20 transgenic (hCD20 Tg) mice and also EL4 and B16 mouse tumour cells expressing human CD20 (hCD20). Importantly, in a hCD20 Tg mouse model, [SCT × Fab'] administered systemically were able to retarget activated OT-I cells to deplete normal B cells, and their performance matched that of a bispecific antibody (BsAb) comprising anti-CD3 and anti-CD20. [SCT × Fab'] were also active therapeutically in an EL4 tumour model. Furthermore, measurement of serum cytokine levels suggests that [SCT × Fab'] are associated with a lower level of inflammatory cytokine release than the BsAb and so may be advantageous clinically in terms of reduced toxicity.


Subject(s)
Antibodies, Bispecific/immunology , Cytotoxicity, Immunologic , Histocompatibility Antigens Class I/immunology , Immunoconjugates/immunology , Neoplasms/immunology , Peptides/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Bispecific/genetics , Antigens, CD20/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Line, Tumor , Disease Models, Animal , Gene Order , Histocompatibility Antigens Class I/genetics , Humans , Immunoconjugates/administration & dosage , Lymphocyte Activation/immunology , Lymphocyte Depletion , Mice , Neoplasms/drug therapy , Neoplasms/mortality , Ovalbumin/immunology , Peptides/chemistry , Protein Binding , Recombinant Fusion Proteins , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/immunology , T-Lymphocytes, Cytotoxic/immunology
15.
Proc Natl Acad Sci U S A ; 107(4): 1654-9, 2010 Jan 26.
Article in English | MEDLINE | ID: mdl-20080692

ABSTRACT

Voltage-gated calcium channels are thought to exist in the plasma membrane as heteromeric proteins, in which the alpha1 subunit is associated with two auxiliary subunits, the intracellular beta subunit and the alpha(2)delta subunit; both of these subunits influence the trafficking and properties of Ca(V)1 and Ca(V)2 channels. The alpha(2)delta subunits have been described as type I transmembrane proteins, because they have an N-terminal signal peptide and a C-terminal hydrophobic and potentially transmembrane region. However, because they have very short C-terminal cytoplasmic domains, we hypothesized that the alpha(2)delta proteins might be associated with the plasma membrane through a glycosylphosphatidylinositol (GPI) anchor attached to delta rather than a transmembrane domain. Here, we provide biochemical, immunocytochemical, and mutational evidence to show that all of the alpha(2)delta subunits studied, alpha(2)delta-1, alpha(2)delta-2, and alpha(2)delta-3, show all of the properties expected of GPI-anchored proteins, both when heterologously expressed and in native tissues. They are substrates for prokaryotic phosphatidylinositol-phospholipase C (PI-PLC) and trypanosomal GPI-PLC, which release the alpha(2)delta proteins from membranes and intact cells and expose a cross-reacting determinant epitope. PI-PLC does not affect control transmembrane or membrane-associated proteins. Furthermore, mutation of the predicted GPI-anchor sites markedly reduced plasma membrane and detergent-resistant membrane localization of alpha(2)delta subunits. We also show that GPI anchoring of alpha(2)delta subunits is necessary for their function to enhance calcium currents, and PI-PLC treatment only reduces calcium current density when alpha(2)delta subunits are coexpressed. In conclusion, this study redefines our understanding of alpha(2)delta subunits, both in terms of their role in calcium-channel function and other roles in synaptogenesis.


Subject(s)
Calcium Channels/metabolism , Glycosylphosphatidylinositols/metabolism , Protein Processing, Post-Translational , Amino Acid Sequence , Animals , COS Cells , Calcium Channels/chemistry , Calcium Channels/genetics , Calcium Channels, L-Type , Chlorocebus aethiops , Mice , Molecular Sequence Data , Mutation , Protein Binding , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Rats
16.
J Neurosci ; 29(13): 4076-88, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19339603

ABSTRACT

Neuropathic pain results from damage to the peripheral sensory nervous system, which may have a number of causes. The calcium channel subunit alpha(2)delta-1 is upregulated in dorsal root ganglion (DRG) neurons in several animal models of neuropathic pain, and this is causally related to the onset of allodynia, in which a non-noxious stimulus becomes painful. The therapeutic drugs gabapentin and pregabalin (PGB), which are both alpha(2)delta ligands, have antiallodynic effects, but their mechanism of action has remained elusive. To investigate this, we used an in vivo rat model of neuropathy, unilateral lumbar spinal nerve ligation (SNL), to characterize the distribution of alpha(2)delta-1 in DRG neurons, both at the light- and electron-microscopic level. We found that, on the side of the ligation, alpha(2)delta-1 was increased in the endoplasmic reticulum of DRG somata, in intracellular vesicular structures within their axons, and in the plasma membrane of their presynaptic terminals in superficial layers of the dorsal horn. Chronic PGB treatment of SNL animals, at a dose that alleviated allodynia, markedly reduced the elevation of alpha(2)delta-1 in the spinal cord and ascending axon tracts. In contrast, it had no effect on the upregulation of alpha(2)delta-1 mRNA and protein in DRGs. In vitro, PGB reduced plasma membrane expression of alpha(2)delta-1 without affecting endocytosis. We conclude that the antiallodynic effect of PGB in vivo is associated with impaired anterograde trafficking of alpha(2)delta-1, resulting in its decrease in presynaptic terminals, which would reduce neurotransmitter release and spinal sensitization, an important factor in the maintenance of neuropathic pain.


Subject(s)
Anticonvulsants/therapeutic use , Neuralgia/pathology , Presynaptic Terminals/metabolism , gamma-Aminobutyric Acid/analogs & derivatives , Analysis of Variance , Animals , Behavior, Animal/drug effects , Calcium Channels/metabolism , Calcium Channels, L-Type , Disease Models, Animal , Endocytosis/drug effects , Functional Laterality , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/ultrastructure , Male , Microscopy, Electron, Transmission/methods , Neuralgia/drug therapy , Pain Measurement/methods , Pregabalin , Presynaptic Terminals/drug effects , Presynaptic Terminals/ultrastructure , Protein Transport/drug effects , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Time Factors , Up-Regulation/drug effects , Up-Regulation/physiology , gamma-Aminobutyric Acid/therapeutic use
17.
Trends Pharmacol Sci ; 28(5): 220-8, 2007 May.
Article in English | MEDLINE | ID: mdl-17403543

ABSTRACT

In this review, we examine what is known about the mechanism of action of the auxiliary alpha2delta subunits of voltage-gated Ca(2+) (Ca(v)) channels. First, to provide some background on the alpha2delta proteins, we discuss the genes encoding these channels, in addition to the topology and predicted structure of the alpha2delta subunits. We then describe the effects of alpha2delta subunits on the biophysical properties of Ca(v) channels and their physiological function. All alpha2delta subunits increase the density at the plasma membrane of Ca(2+) channels activated by high voltage, and we discuss what is known about the mechanism underlying this trafficking. Finally, we consider the link between alpha2delta subunits and disease, both in terms of spontaneous and engineered mouse mutants that show cerebellar ataxia and spike-wave epilepsy, and in terms of neuropathic pain and the mechanism of action of the gabapentinoid drugs - small-molecule ligands of the alpha2delta-1 and alpha2delta-2 subunits.


Subject(s)
Calcium Channels/physiology , Ion Channel Gating , Protein Subunits/physiology , Amines/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels/genetics , Calcium Channels/ultrastructure , Cell Membrane/metabolism , Cerebellar Ataxia/physiopathology , Cyclohexanecarboxylic Acids/pharmacology , Epilepsy/physiopathology , Gabapentin , Humans , Mice , Neuralgia/physiopathology , gamma-Aminobutyric Acid/pharmacology
18.
J Neurosci ; 26(34): 8748-57, 2006 Aug 23.
Article in English | MEDLINE | ID: mdl-16928863

ABSTRACT

The accessory alpha2delta subunits of voltage-gated calcium channels are highly glycosylated transmembrane proteins that interact with calcium channel alpha1 subunits to enhance calcium currents. We compared the membrane localization and processing of native cerebellar alpha2delta-2 subunits with alpha2delta-2 stably expressed in tsA-201 cells. We identified that alpha2delta-2 is completely concentrated in cholesterol-rich microdomains (lipid rafts) in cerebellum, in which it substantially colocalizes with the calcium channel alpha1 subunit CaV2.1, although CaV2.1 is also present in the Triton X-100-soluble fraction. In tsA-201 cells, unlike cerebellum, alpha2delta-2 is not completely proteolytically processed into alpha2-2 and delta-2. However, this processing is more complete in the lipid raft fraction of tsA-201 cells, in which alpha2delta-2 also colocalizes with CaV2.1. Cholesterol depletion of intact cells disrupted their lipid rafts and enhanced CaV2.1/alpha2delta-2/beta4 currents. Furthermore, alpha2delta-2 coimmunoprecipitates with lipid raft-associated proteins of the stomatin family. The apparent affinity of alpha2delta-2 for its ligand gabapentin is increased markedly in the cholesterol-rich microdomain fractions, in both cerebellum and the stable alpha2delta-2 cell line. In contrast, alpha2delta-2 containing a point mutation (R282A) has a much lower affinity for gabapentin, and this is not enhanced in the lipid raft fraction. This R282A mutant alpha2delta-2 shows reduced functionality in terms of enhancement of CaV2.1/beta4 calcium currents, suggesting that the integrity of the gabapentin binding site may be important for normal functioning of alpha2delta-2. Together, these results indicate that both alpha2delta-2 and CaV2.1 are normally associated with cholesterol-rich microdomains, and this influences their functionality.


Subject(s)
Calcium Channels, N-Type/metabolism , Calcium Channels/metabolism , Cerebellum/metabolism , Membrane Microdomains/metabolism , Alanine , Amines/antagonists & inhibitors , Amines/metabolism , Amines/pharmacology , Animals , Arginine , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Calcium Channels/genetics , Calcium Channels/physiology , Calcium Channels, N-Type/physiology , Cell Line , Cholesterol/metabolism , Cyclohexanecarboxylic Acids/antagonists & inhibitors , Cyclohexanecarboxylic Acids/metabolism , Cyclohexanecarboxylic Acids/pharmacology , Electric Conductivity , Gabapentin , Immunoprecipitation , Mice , Mutation/physiology , Nerve Tissue Proteins/metabolism , Purkinje Cells/metabolism , Tissue Distribution , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology
19.
J Biomol Screen ; 9(5): 391-7, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15296638

ABSTRACT

STK15/Aurora2 is a centrosome-associated serine/threonine kinase, the protein levels and kinase activity of which rise during G2 and mitosis. STK15 overexpression induces tumorigenesis and is amplified in various human cancers and tumor cell lines. Thus, STK15 represents an important therapeutic target for small molecule inhibitors that would disrupt its activity and block cell proliferation. The availability of a robust and selective small molecule inhibitor would also provide a useful tool for identification of the potential role of STK15 in cell cycle regulation and tumor development. The authors report the development of a novel, fast, simple microplate assay for STK15 activity suitable for high-throughput screening. In the assay, gamma-(33)P-ATP and STK15 were incubated in a myelin basic protein (MBP)-coated FlashPlate(R) to generate a scintillation signal. The assay was reproducible, the signal-to-noise ratio was high (11) and the Z' factor was 0.69. The assay was easily adapted to a robotic system for drug discovery programs targeting STK15. The authors also demonstrate that STK15 is regulated by phosphorylation and the N-amino terminal domain of the protein. Treatment with phosphatase inhibitors (okadaic acid) or deletion of the N-amino terminal domain results in a significant increase in the enzymatic activity.


Subject(s)
Enzyme Inhibitors/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Amino Acid Sequence , Aurora Kinase A , Aurora Kinases , Enzyme Inhibitors/pharmacology , Molecular Sequence Data
20.
Farm. al día ; 3(7): 372-3, 1992. ilus
Article in Spanish | LILACS | ID: lil-270095

Subject(s)
Humans , Male , Head , Pain/complications , Men
SELECTION OF CITATIONS
SEARCH DETAIL
...