Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Neurobiol Dis ; 134: 104683, 2020 02.
Article in English | MEDLINE | ID: mdl-31765727

ABSTRACT

Repeated mild traumatic brain injury (rmTBI) can lead to development of chronic traumatic encephalopathy (CTE), which is characterized by progressive neurodegeneration with presence of white matter damage, gliosis and hyper-phosphorylated tau. While animal models of rmTBI have been documented, few characterize the molecular pathogenesis and expression profiles of relevant injured brain regions. Additionally, while the usage of transgenic tau mice in rmTBI is prevalent, the effects of tau on pathological outcomes has not been well studied. Here we characterized a 42-impact closed-head rmTBI paradigm on 3-4Ā month old male C57BL/6 (WT) and Tau-overexpressing mice (Tau58.4). This injury paradigm resulted in chronic gliosis, T-cell infiltration, and demyelination of the optic nerve and associated white matter tracts at 1-month post-injury. At 3-months post-injury, Tau58.4 mice showed progressive neuroinflammation and neurodegeneration in multiple brain regions compared to WT mice. Corresponding to histopathology, RNAseq of the optic nerve tract at 1-month post-injury showed significant upregulation of inflammatory pathways and downregulation of myelin synthetic pathways in both genotypes. However, Tau58.4 mice showed additional changes in neurite development, protein processing, and cell stress. Comparisons with published transcriptomes of human Alzheimer's Disease and CTE revealed common signatures including neuroinflammation and downregulation of protein phosphatases. We next investigated the demyelination and T-cell infiltration phenotypes to determine whether these offer potential avenues for therapeutic intervention. Tau58.4 mice were treated with the histamine H3 receptor antagonist GSK239512 for 1-month post-injury to promote remyelination of white matter lesions. This restored myelin gene expression to sham levels but failed to repair the histopathologic lesions. Likewise, injured T-cell-deficient Rag2/Il2rg (R2G2) mice also showed evidence for inflammation and loss of myelin. However, unlike immune-competent mice, R2G2 mice had altered myeloid cell gene expression and fewer demyelinated lesions. Together this data shows that rmTBI leads to chronic white matter inflammatory demyelination and axonal loss exacerbated by human tau overexpression but suggests that immune-suppression and remyelination alone are insufficient to reverse damage.


Subject(s)
Brain Concussion/metabolism , Brain Concussion/pathology , Brain/metabolism , Brain/pathology , tau Proteins/metabolism , Animals , Brain Concussion/complications , Encephalitis/complications , Encephalitis/metabolism , Encephalitis/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , White Matter/metabolism , White Matter/pathology
2.
J Neurosci ; 33(6): 2457-64, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23392674

ABSTRACT

Accumulation of Ɵ-amyloid (AƟ) in the brain is believed to contribute to the pathology of Alzheimer's Disease (AD). AƟ levels are controlled by the production of AƟ from amyloid precursor protein, degradation by proteases, and peripheral clearance. In this study we sought to determine whether enhancing clearance of plasma AƟ with a peripherally administered AƟ-degrading protease would reduce brain AƟ levels through a peripheral sink. Neprilysin (NEP) is a zinc-dependent metalloprotease that is one of the key AƟ-degrading enzymes in the brain. We developed a NEP fusion protein with in vitro degradation of AƟ and a 10 day plasma half-life in mouse. Intravenous administration of NEP to wild-type and APP23 transgenic mice resulted in dose-dependent clearance of plasma AƟ. However, this did not correspond to reduced levels of soluble brain AƟ with treatment up to 5 weeks in WT mice or formic acid-extractable brain AƟ with 3 month treatment in aged APP23. In contrast, intracranial injection of NEP resulted in an acute decrease in soluble brain AƟ. We found no change in amyloid precursor protein gene expression in mice treated with intravenous NEP, suggesting that the lack of effects in the brain following this route of administration was not caused by compensatory upregulation of AƟ production. Taken together, these results suggest a lack of a robust peripheral AƟ efflux sink through which brain amyloid burdens can be therapeutically reduced.


Subject(s)
Amyloid beta-Peptides/blood , Amyloid beta-Protein Precursor/blood , Brain/drug effects , Brain/metabolism , Neprilysin/pharmacology , Proteolysis/drug effects , Amino Acid Sequence , Animals , Brain/pathology , Dose-Response Relationship, Drug , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data
3.
Front Neuroimaging ; 3: 1356713, 2024.
Article in English | MEDLINE | ID: mdl-38783990

ABSTRACT

Purpose: To test the ability of inversion-recovery ultrashort echo time (IR-UTE) MRI to directly detect demyelination in mice using a standard cuprizone mouse model. Methods: Non-aqueous myelin protons have ultrashort T2s and are "invisible" with conventional MRI sequences but can be detected with UTE sequences. The IR-UTE sequence uses an adiabatic inversion-recovery preparation to suppress the long T2 water signal so that the remaining signal is from the ultrashort T2 myelin component. In this study, eight 8-week-old C57BL/6 mice were fed cuprizone (n = 4) or control chow (n = 4) for 5 weeks and then imaged by 3D IR-UTE MRI. The differences in IR-UTE signal were compared in the major white matter tracts in the brain and correlated with the Luxol Fast Blue histochemical marker of myelin. Results: IR-UTE signal decreased in cuprizone-treated mice in white matter known to be sensitive to demyelination in this model, such as the corpus callosum, but not in white matter known to be resistant to demyelination, such as the internal capsule. These findings correlated with histochemical staining of myelin content. Conclusions: 3D IR-UTE MRI was sensitive to cuprizone-induced demyelination in the mouse brain, and is a promising noninvasive method for measuring brain myelin content.

4.
Biochem J ; 426(2): 219-28, 2010 Feb 09.
Article in English | MEDLINE | ID: mdl-20015050

ABSTRACT

HGF (hepatocyte growth factor) is a pleiotropic cytokine homologous to the serine protease zymogen plasminogen that requires canonical proteolytic cleavage to gain functional activity. The activating proteases are key components of its regulation, but controversy surrounds their identity. Using quantitative analysis we found no evidence for activation by uPA (urokinase plasminogen activator), despite reports that this is a principal activator of pro-HGF. This was unaffected by a wide range of experimental conditions, including the use of various molecular forms of both HGF and uPA, and the presence of uPAR (uPA receptor) or heparin. In contrast the catalytic domains of the TTSPs (type-II transmembrane serine proteases) matriptase and hepsin were highly efficient activators (50% activation at 0.1 and 3.4 nM respectively), at least four orders of magnitude more efficient than uPA. PS-SCL (positional-scanning synthetic combinatorial peptide libraries) were used to identify consensus sequences for the TTSPs, which in the case of hepsin corresponded to the pro-HGF activation sequence, demonstrating a high specificity for this reaction. Both TTSPs were also found to be efficient activators at the cell surface. Activation of pro-HGF by PC3 prostate carcinoma cells was abolished by both protease inhibition and matriptase-targeting siRNA (small interfering RNA), and scattering of MDCK (Madin-Darby canine kidney) cells in the presence of pro-HGF was abolished by inhibition of matriptase. Hepsin-transfected HEK (human embryonic kidney)-293 cells also activated pro-HGF. These observations demonstrate that, in contrast with the uPA/uPAR system, the TTSPs matriptase and hepsin are direct pericellular activators of pro-HGF, and that together these proteins may form a pathway contributing to their involvement in pathological situations, including cancer.


Subject(s)
Cell Membrane/enzymology , Hepatocyte Growth Factor/metabolism , Protein Processing, Post-Translational , Serine Endopeptidases/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Animals , Cell Line, Tumor , Cell Membrane/genetics , Dogs , Hepatocyte Growth Factor/genetics , Humans , Serine Endopeptidases/genetics , Urokinase-Type Plasminogen Activator/genetics
5.
ChemMedChem ; 15(16): 1562-1570, 2020 08 19.
Article in English | MEDLINE | ID: mdl-32613743

ABSTRACT

Loss of Ɵ-cell mass and function can lead to insufficient insulin levels and ultimately to hyperglycemia and diabetes mellitus. The mainstream treatment approach involves regulation of insulin levels; however, approaches intended to increase Ɵ-cell mass are less developed. Promoting Ɵ-cell proliferation with low-molecular-weight inhibitors of dual-specificity tyrosine-regulated kinase 1A (DYRK1A) offers the potential to treat diabetes with oral therapies by restoring Ɵ-cell mass, insulin content and glycemic control. GNF4877, a potent dual inhibitor of DYRK1A and glycogen synthase kinase 3Ɵ (GSK3Ɵ) was previously reported to induce primary human Ɵ-cell proliferation inĆ¢Ā€Ā…vitro and inĆ¢Ā€Ā…vivo. Herein, we describe the lead optimization that lead to the identification of GNF4877 from an aminopyrazine hit identified in a phenotypic high-throughput screening campaign measuring Ɵ-cell proliferation.


Subject(s)
Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Insulin-Secreting Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Mice , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Rats , Structure-Activity Relationship , Dyrk Kinases
6.
J Med Chem ; 63(6): 2958-2973, 2020 03 26.
Article in English | MEDLINE | ID: mdl-32077280

ABSTRACT

Autoimmune deficiency and destruction in either Ɵ-cell mass or function can cause insufficient insulin levels and, as a result, hyperglycemia and diabetes. Thus, promoting Ɵ-cell proliferation could be one approach toward diabetes intervention. In this report we describe the discovery of a potent and selective DYRK1A inhibitor GNF2133, which was identified through optimization of a 6-azaindole screening hit. In vitro, GNF2133 is able to proliferate both rodent and human Ɵ-cells. In vivo, GNF2133 demonstrated significant dose-dependent glucose disposal capacity and insulin secretion in response to glucose-potentiated arginine-induced insulin secretion (GPAIS) challenge in rat insulin promoter and diphtheria toxin A (RIP-DTA) mice. The work described here provides new avenues to disease altering therapeutic interventions in the treatment of type 1 diabetes (T1D).


Subject(s)
Aza Compounds/chemistry , Aza Compounds/pharmacology , Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Indoles/chemistry , Indoles/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Aza Compounds/pharmacokinetics , Cell Proliferation/drug effects , Cells, Cultured , Diabetes Mellitus, Type 1/metabolism , Humans , Hypoglycemic Agents/pharmacokinetics , Indoles/pharmacokinetics , Insulin Secretion/drug effects , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Mice , Molecular Docking Simulation , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Dyrk Kinases
7.
Bioorg Med Chem Lett ; 18(22): 5895-9, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18752942

ABSTRACT

Structure-based design was utilized to guide the early stage optimization of a substrate-like inhibitor to afford potent peptidomimetic inhibitors of the channel-activating protease prostasin. The first X-ray crystal structures of prostasin with small molecule inhibitors bound to the active site are also reported.


Subject(s)
Serine Endopeptidases/drug effects , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/pharmacology , Combinatorial Chemistry Techniques , Crystallography, X-Ray , Molecular Mimicry , Molecular Structure , Protein Conformation , Serine Proteinase Inhibitors/chemistry , Structure-Activity Relationship
8.
ACS Chem Biol ; 12(12): 3126-3133, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29099173

ABSTRACT

Glycosaminoglycans (GAGs) play critical roles in diverse processes ranging from viral infection to neuroregeneration. Their regiospecific sulfation patterns, which are generated by sulfotransferases, are key structural determinants that underlie their biological activity. Small-molecule modulators of these sulfotransferases could serve as powerful tools for understanding the physiological functions of GAGs, as well as potential therapeutic leads for human diseases. Here, we report the development of the first cell-permeable, small-molecule inhibitor selective for GAG sulfotransferases, which was obtained using a high-throughput screen targeted against Chst15, the sulfotransferase responsible for biosynthesis of chondroitin sulfate-E (CS-E). We demonstrate that the molecule specifically inhibits GAG sulfotransferases in vitro, decreases CS-E and overall sulfation levels on cell-surface and secreted chondroitin sulfate proteoglycans (CSPGs), and reverses CSPG-mediated inhibition of axonal growth. These studies pave the way toward a new set of pharmacological tools for interrogating GAG sulfation-dependent processes and may represent a novel therapeutic approach for neuroregeneration.


Subject(s)
Gene Expression Regulation, Enzymologic/drug effects , Glycosaminoglycans/chemistry , Glycosaminoglycans/metabolism , Membrane Glycoproteins/metabolism , Sulfotransferases/metabolism , Animals , Cloning, Molecular , High-Throughput Screening Assays , Membrane Glycoproteins/genetics , Mice , Microsomes/metabolism , NIH 3T3 Cells , Rats , Sulfotransferases/genetics
9.
Biochim Biophys Acta ; 1600(1-2): 128-37, 2002 Nov 04.
Article in English | MEDLINE | ID: mdl-12445468

ABSTRACT

Death-associated protein kinase (DAPK) is a pro-apoptotic, calmodulin (CaM)-regulated protein kinase whose mRNA levels increase following cerebral ischemia. However, the relationship between DAPK catalytic activity and cerebral ischemia is not known. This knowledge is critical as DAPK function is dependent on the catalytic activity of its kinase domain. Consequently, we examined DAPK catalytic activity in a rat model of neonatal cerebral hypoxia-ischemia (HI). An increase in DAPK specific activity was found in homogenates of the hippocampus from the injured right hemisphere, compared to the uninjured left hemisphere, 7 days after injury. The results raised the possibility that an upregulation of DAPK activity might be associated with the recovery phase of HI, during which neuronal repair and differentiation are initiated. Therefore, we examined the change of DAPK in an experimentally tractable cell culture model of neuronal differentiation. We found that DAPK catalytic activity and protein levels increase after nerve growth factor (NGF)-induced differentiation of rat PC12 cells. These results suggest that DAPK may have a previously unappreciated role in neuronal development or recovery from injury, and that potential future therapies targeting DAPK should consider a restricted time window.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Hippocampus/enzymology , Hypoxia-Ischemia, Brain/enzymology , Animals , Animals, Newborn , Apoptosis Regulatory Proteins , Binding Sites , Brain/enzymology , Brain/pathology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Catalysis , Cell Differentiation , Death-Associated Protein Kinases , Enzyme Activation , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Hypoxia-Ischemia, Brain/diagnosis , Hypoxia-Ischemia, Brain/pathology , Kinetics , Models, Molecular , Nerve Growth Factor/pharmacology , PC12 Cells , Rats , Rats, Wistar , Up-Regulation
10.
Pharmacol Ther ; 93(2-3): 217-24, 2002.
Article in English | MEDLINE | ID: mdl-12191613

ABSTRACT

Death-associated protein kinase (DAPK) is a calmodulin-regulated serine/threonine protein kinase associated with neuronal cell death in animal models of disease. The recent determination of the 1.5A crystal structure of the catalytic kinase domain of DAPK, the discovery of amino acid sequence motifs with sites that are preferentially phosphorylated by this kinase, and the development of a quantitative enzyme activity assay provide a firm foundation for future studies into its regulation, the identification of its physiological substrates, and discovery of inhibitors. We summarize the relevant background and ongoing investigations that will increase our understanding of the role and regulation of this prototype death-associated kinase.


Subject(s)
Apoptosis/physiology , Calcium-Calmodulin-Dependent Protein Kinases , Animals , Apoptosis Regulatory Proteins , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Death-Associated Protein Kinases
11.
Elife ; 42015 May 22.
Article in English | MEDLINE | ID: mdl-26001275

ABSTRACT

Piezo ion channels are activated by various types of mechanical stimuli and function as biological pressure sensors in both vertebrates and invertebrates. To date, mechanical stimuli are the only means to activate Piezo ion channels and whether other modes of activation exist is not known. In this study, we screened ~3.25 million compounds using a cell-based fluorescence assay and identified a synthetic small molecule we termed Yoda1 that acts as an agonist for both human and mouse Piezo1. Functional studies in cells revealed that Yoda1 affects the sensitivity and the inactivation kinetics of mechanically induced responses. Characterization of Yoda1 in artificial droplet lipid bilayers showed that Yoda1 activates purified Piezo1 channels in the absence of other cellular components. Our studies demonstrate that Piezo1 is amenable to chemical activation and raise the possibility that endogenous Piezo1 agonists might exist. Yoda1 will serve as a key tool compound to study Piezo1 regulation and function.


Subject(s)
Ion Channels/agonists , Ion Channels/metabolism , Mechanotransduction, Cellular/physiology , Small Molecule Libraries/pharmacology , Animals , Fluorescence , High-Throughput Screening Assays , Humans , Mice
12.
PLoS One ; 10(6): e0127498, 2015.
Article in English | MEDLINE | ID: mdl-26098886

ABSTRACT

Englerin A is a structurally unique natural product reported to selectively inhibit growth of renal cell carcinoma cell lines. A large scale phenotypic cell profiling experiment (CLiP) of englerin A on Ā¬over 500 well characterized cancer cell lines showed that englerin A inhibits growth of a subset of tumor cell lines from many lineages, not just renal cell carcinomas. Expression of the TRPC4 cation channel was the cell line feature that best correlated with sensitivity to englerin A, suggesting the hypothesis that TRPC4 is the efficacy target for englerin A. Genetic experiments demonstrate that TRPC4 expression is both necessary and sufficient for englerin A induced growth inhibition. Englerin A induces calcium influx and membrane depolarization in cells expressing high levels of TRPC4 or its close ortholog TRPC5. Electrophysiology experiments confirmed that englerin A is a TRPC4 agonist. Both the englerin A induced current and the englerin A induced growth inhibition can be blocked by the TRPC4/C5 inhibitor ML204. These experiments confirm that activation of TRPC4/C5 channels inhibits tumor cell line proliferation and confirms the TRPC4 target hypothesis generated by the cell line profiling. In selectivity assays englerin A weakly inhibits TRPA1, TRPV3/V4, and TRPM8 which suggests that englerin A may bind a common feature of TRP ion channels. In vivo experiments show that englerin A is lethal in rodents near doses needed to activate the TRPC4 channel. This toxicity suggests that englerin A itself is probably unsuitable for further drug development. However, since englerin A can be synthesized in the laboratory, it may be a useful chemical starting point to identify novel modulators of other TRP family channels.


Subject(s)
Cell Proliferation/drug effects , Sesquiterpenes, Guaiane/pharmacology , TRPC Cation Channels/agonists , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/drug therapy , Cell Line, Tumor , HEK293 Cells , Humans , Indoles/pharmacology , Kidney Neoplasms/drug therapy , Mice , Mice, Nude , Piperidines/pharmacology , RNA Interference , RNA, Small Interfering , Rats , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/genetics , Transfection
13.
J Med Chem ; 57(12): 5129-40, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24884675

ABSTRACT

A high throughput screening campaign identified 5-(2-chlorophenyl)indazole compound 4 as an antagonist of the transient receptor potential A1 (TRPA1) ion channel with IC50 = 1.23 ĀµM. Hit to lead medicinal chemistry optimization established the SAR around the indazole ring system, demonstrating that a trifluoromethyl group at the 2-position of the phenyl ring in combination with various substituents at the 6-position of the indazole ring greatly contributed to improvements in vitro activity. Further lead optimization resulted in the identification of compound 31, a potent and selective antagonist of TRPA1 in vitro (IC50 = 0.015 ĀµM), which has moderate oral bioavailability in rodents and demonstrates robust activity in vivo in several rodent models of inflammatory pain.


Subject(s)
Indazoles/chemistry , Nerve Tissue Proteins/antagonists & inhibitors , Transient Receptor Potential Channels/antagonists & inhibitors , Administration, Oral , Analgesics/chemistry , Analgesics/pharmacokinetics , Analgesics/pharmacology , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/pharmacology , Biological Availability , CHO Cells , Calcium Channels , Cricetulus , Freund's Adjuvant , Humans , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Indazoles/pharmacokinetics , Indazoles/pharmacology , Male , Mice, Inbred C57BL , Mustard Plant , Plant Oils , Rats, Wistar , Species Specificity , Structure-Activity Relationship , TRPA1 Cation Channel , TRPC Cation Channels/antagonists & inhibitors
14.
Biochemistry ; 45(45): 13614-21, 2006 Nov 14.
Article in English | MEDLINE | ID: mdl-17087515

ABSTRACT

Death-associated protein kinase (DAPK) is a pro-apoptotic, calcium/calmodulin-regulated protein kinase that is a drug discovery target for neurodegenerative disorders. Despite the potential profound physiological role of DAPK in neuronal function and pathophysiology, the endogenous substrate(s) of this kinase and the mechanisms via which DAPK elicits its biological action remain largely unknown. We report here that the mammalian 40S ribosomal protein S6 is a DAPK substrate. Results from immunoprecipitation experiments are consistent with endogenous DAPK being associated with endogenous S6 in rat brain. When S6 is a component of the 40S ribosomal subunit complex, DAPK selectively phosphorylates it at serine 235, one of the five sites in S6 that are phosphorylated by the S6 kinase family of proteins. The amino acid sequence flanking serine 235 matches the established pattern for DAPK peptide and protein substrates. Kinetic analyses using purified 40S subunits revealed a K(m) value of 9 microM, consistent with S6 being a potential physiological substrate of DAPK. This enzyme-substrate relationship has functional significance. DAPK suppresses translation in rabbit reticulocyte lysate, and treatment of neuroblastoma cells with a stimulator of DAPK reduces protein synthesis. In both cases, suppression of translation correlates with increased phosphorylation of S6 at serine 235. These results demonstrate that DAPK is a S6 kinase and provide evidence for a novel role of DAPK in the regulation of translation.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Brain/enzymology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Protein Biosynthesis/drug effects , Ribosomal Protein S6/metabolism , Amino Acid Sequence , Animals , Death-Associated Protein Kinases , Ethinyl Estradiol/metabolism , Megestrol Acetate/metabolism , Phosphorylation , Rats , Ribosomal Protein S6 Kinases, 70-kDa/metabolism
15.
Expert Opin Ther Targets ; 6(4): 497-506, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12223064

ABSTRACT

Death associated protein kinase (DAPK) is a calmodulin (CaM)-regulated serine/threonine protein kinase implicated in diverse apoptosis pathways, including those involved in neuronal cell death and tumour suppression. The requirement of DAPK catalytic activity for its proposed cell functions and the validation of protein kinases as therapeutic targets demand that DAPK be examined as a potential therapeutic target in human disease. The relevant placement of DAPK activity in apoptosis pathways is at an early stage of investigation, making its study as a therapeutic target tenuous. However, the current body of knowledge raises the possibility of DAPK as a therapeutic target for diseases characterised by rapid neurodegeneration, such as stroke or traumatic brain injury. The unmet need in these diseases is for an acute treatment schedule that might reduce neuronal loss. Bioavailable inhibitors of DAPK catalytic activity that target the central nervous system have a potential to fill this need. The development of such DAPK inhibitors is now feasible based on the recent emergence of enabling technology and knowledge. These include a quantitative and selective enzyme assay, a high resolution structure of the active catalytic domain and discovery of cell-permeable, low molecular weight inhibitors of CaM kinases that cross the blood-brain barrier. DAPK as a potential therapeutic target for cancer is less attractive due to the incomplete state of knowledge about DAPK and inherent limitations in drug development for the discovery of specific activators of genes downregulated by promoter hypermethylation. This article provides a brief summary of relevant research and the rationale that is at the foundation of this opinion.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Drug Design , Protein Kinase Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Blood-Brain Barrier , Brain Ischemia/drug therapy , Brain Ischemia/enzymology , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Calmodulin/physiology , Central Nervous System Agents/pharmacology , Central Nervous System Agents/therapeutic use , Ceramides/metabolism , Death-Associated Protein Kinases , Dogs , Drug Evaluation, Preclinical , Enzyme Induction/drug effects , Genes, Tumor Suppressor , Humans , Mice , Models, Animal , Models, Molecular , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/physiology , Neoplasms/drug therapy , Neoplasms/enzymology , Nerve Degeneration/drug therapy , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/physiology , Protein Conformation , Protein Kinase Inhibitors/therapeutic use
16.
Biochemistry ; 43(25): 8116-24, 2004 Jun 29.
Article in English | MEDLINE | ID: mdl-15209507

ABSTRACT

Death-associated protein kinase (DAPK) is a calmodulin (CaM)-regulated protein kinase and a drug-discovery target for neurodegenerative diseases. However, a protein substrate relevant to neuronal death had not been described. We identified human brain CaM-regulated protein kinase kinase (CaMKK), an enzyme key to neuronal survival, as the first relevant substrate protein by using a focused proteomics- and informatics-based approach that can be generalized to protein kinase open reading frames identified in genome projects without prior knowledge of biochemical context. First, DAPK-interacting proteins were detected in yeast two-hybrid screens and in immunoprecipitates of brain extracts. Second, potential phosphorylation site sequences in yeast two-hybrid hits were identified on the basis of our previous results from positional-scanning synthetic-peptide substrate libraries and molecular modeling. Third, reconstitution assays using purified components demonstrated that DAPK phosphorylates CaMKK with a stoichiometry of nearly 1 mol of phosphate per mole of CaMKK and a K(m) value of 3 microM. Fourth, S511 was identified as the phosphorylation site by peptide mapping using mass spectrometry, site-directed mutagenesis, and Western blot analysis with a site-directed antisera targeting the phosphorylated sequence. Fifth, a potential mechanism of action was identified on the basis of the location of S511 near the CaM recognition domain of CaMKK and demonstrated by attenuation of CaM-stimulated CaMKK autophosphorylation after DAPK phosphorylation. The results raise the possibility of a CaM-regulated protein kinase cascade as a key mechanism in acute neurodegeneration amenable to therapeutic targeting.


Subject(s)
Brain/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Apoptosis Regulatory Proteins , Binding Sites , Brain/cytology , Brain/enzymology , Calcium-Calmodulin-Dependent Protein Kinase Kinase , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Calmodulin/metabolism , Cell Survival/physiology , Death-Associated Protein Kinases , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Neurons/cytology , Neurons/enzymology , Phosphorylation , Precipitin Tests , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Two-Hybrid System Techniques , Yeasts/genetics
17.
Bioorg Med Chem Lett ; 13(20): 3465-70, 2003 Oct 20.
Article in English | MEDLINE | ID: mdl-14505650

ABSTRACT

Death associated protein kinase (DAPK) is a calcium and calmodulin regulated enzyme that functions early in eukaryotic programmed cell death, or apoptosis. To validate DAPK as a potential drug discovery target for acute brain injury, the first small molecule DAPK inhibitor was synthesized and tested in vivo. A single injection of the aminopyridazine-based inhibitor administered 6 h after injury attenuated brain tissue or neuronal biomarker loss measured, respectively, 1 week and 3 days later. Because aminopyridazine is a privileged structure in neuropharmacology, we determined the high-resolution crystal structure of a binary complex between the kinase domain and a molecular fragment of the DAPK inhibitor. The co-crystal structure describes a structural basis for interaction and provides a firm foundation for structure-assisted design of lead compounds with appropriate molecular properties for future drug development.


Subject(s)
Apoptosis , Brain Injuries/prevention & control , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hypoxia-Ischemia, Brain/prevention & control , Pyridazines/pharmacology , Animals , Apoptosis Regulatory Proteins , Brain Injuries/etiology , Death-Associated Protein Kinases , Enzyme Inhibitors/chemistry , Hypoxia-Ischemia, Brain/complications , Mice , Pyridazines/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL