Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Neuropathol Commun ; 6(1): 37, 2018 05 07.
Article in English | MEDLINE | ID: mdl-29730992

ABSTRACT

In Alzheimer's disease (AD) and related tauopathies, the microtubule-associated protein tau is highly phosphorylated and aggregates to form neurofibrillary tangles that are characteristic of these neurodegenerative diseases. Our previous work has demonstrated that the thousand-and-one amino acid kinases (TAOKs) 1 and 2 phosphorylate tau on more than 40 residues in vitro. Here we show that TAOKs are phosphorylated and active in AD brain sections displaying mild (Braak stage II), intermediate (Braak stage IV) and advanced (Braak stage VI) tau pathology and that active TAOKs co-localise with both pre-tangle and tangle structures. TAOK activity is also enriched in pathological tau containing sarkosyl-insoluble extracts prepared from AD brain. Two new phosphorylated tau residues (T123 and T427) were identified in AD brain, which appear to be targeted specifically by TAOKs. A new small molecule TAOK inhibitor (Compound 43) reduced tau phosphorylation on T123 and T427 and also on additional pathological sites (S262/S356 and S202/T205/S208) in vitro and in cell models. The TAOK inhibitor also decreased tau phosphorylation in differentiated primary cortical neurons without affecting markers of synapse and neuron health. Notably, TAOK activity also co-localised with tangles in post-mortem frontotemporal lobar degeneration (FTLD) brain tissue. Furthermore, the TAOK inhibitor decreased tau phosphorylation in induced pluripotent stem cell derived neurons from FTLD patients, as well as cortical neurons from a transgenic mouse model of tauopathy (Tau35 mice). Our results demonstrate that abnormal TAOK activity is present at pre-tangles and tangles in tauopathies and that TAOK inhibition effectively decreases tau phosphorylation on pathological sites. Thus, TAOKs may represent a novel target to reduce or prevent tau-associated neurodegeneration in tauopathies.


Subject(s)
Enzyme Inhibitors/therapeutic use , Nerve Degeneration/drug therapy , Nerve Degeneration/etiology , Protein Serine-Threonine Kinases/metabolism , Tauopathies/complications , tau Proteins/metabolism , Animals , Brain/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cerebral Cortex , Dose-Response Relationship, Drug , Female , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/physiology , L-Lactate Dehydrogenase/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Nerve Degeneration/pathology , Neurons/drug effects , Neurons/metabolism , Phosphorylation/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Transfection
2.
Mol Cancer Ther ; 16(11): 2410-2421, 2017 11.
Article in English | MEDLINE | ID: mdl-28830982

ABSTRACT

Thousand-and-one amino acid kinases (TAOK) 1 and 2 are activated catalytically during mitosis and can contribute to mitotic cell rounding and spindle positioning. Here, we characterize a compound that inhibits TAOK1 and TAOK2 activity with IC50 values of 11 to 15 nmol/L, is ATP-competitive, and targets these kinases selectively. TAOK inhibition or depletion in centrosome-amplified SKBR3 or BT549 breast cancer cell models increases the mitotic population, the percentages of mitotic cells displaying amplified centrosomes and multipolar spindles, induces cell death, and inhibits cell growth. In contrast, nontumorigenic and dividing bipolar MCF-10A breast cells appear less dependent on TAOK activity and can complete mitosis and proliferate in the presence of the TAOK inhibitor. We demonstrate that TAOK1 and TAOK2 localize to the cytoplasm and centrosomes respectively during mitosis. Live cell imaging shows that the TAOK inhibitor prolongs the duration of mitosis in SKBR3 cells, increases mitotic cell death, and reduces the percentages of cells exiting mitosis, whereas MCF-10A cells continue to divide and proliferate. Over 80% of breast cancer tissues display supernumerary centrosomes, and tumor cells frequently cluster extra centrosomes to avoid multipolar mitoses and associated cell death. Consequently, drugs that stimulate centrosome declustering and induce multipolarity are likely to target dividing centrosome-amplified cancer cells preferentially, while sparing normal bipolar cells. Our results demonstrate that TAOK inhibition can enhance centrosome declustering and mitotic catastrophe in cancer cells, and these proteins may therefore offer novel therapeutic targets suitable for drug inhibition and the potential treatment of breast cancers, where supernumerary centrosomes occur. Mol Cancer Ther; 16(11); 2410-21. ©2017 AACR.


Subject(s)
Breast Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Protein Kinases/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Centrosome/drug effects , Female , Humans , Mitosis/drug effects , Protein Kinase Inhibitors/chemical synthesis , Protein Kinases/drug effects , Protein Serine-Threonine Kinases/genetics , Spindle Apparatus/drug effects
3.
J Biol Chem ; 288(21): 15418-29, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23585562

ABSTRACT

In Alzheimer disease (AD), the microtubule-associated protein tau is highly phosphorylated and aggregates into characteristic neurofibrillary tangles. Prostate-derived sterile 20-like kinases (PSKs/TAOKs) 1 and 2, members of the sterile 20 family of kinases, have been shown to regulate microtubule stability and organization. Here we show that tau is a good substrate for PSK1 and PSK2 phosphorylation with mass spectrometric analysis of phosphorylated tau revealing more than 40 tau residues as targets of these kinases. Notably, phosphorylated residues include motifs located within the microtubule-binding repeat domain on tau (Ser-262, Ser-324, and Ser-356), sites that are known to regulate tau-microtubule interactions. PSK catalytic activity is enhanced in the entorhinal cortex and hippocampus, areas of the brain that are most susceptible to Alzheimer pathology, in comparison with the cerebellum, which is relatively spared. Activated PSK is associated with neurofibrillary tangles, dystrophic neurites surrounding neuritic plaques, neuropil threads, and granulovacuolar degeneration bodies in AD brain. By contrast, activated PSKs and phosphorylated tau are rarely detectible in immunostained control human brain. Our results demonstrate that tau is a substrate for PSK and suggest that this family of kinases could contribute to the development of AD pathology and dementia.


Subject(s)
Alzheimer Disease/metabolism , MAP Kinase Kinase Kinases/metabolism , Neurons/metabolism , tau Proteins/metabolism , Alzheimer Disease/pathology , Amino Acid Motifs , Animals , COS Cells , Cerebellum/metabolism , Cerebellum/pathology , Chlorocebus aethiops , Entorhinal Cortex/metabolism , Entorhinal Cortex/pathology , Female , Hippocampus/metabolism , Hippocampus/pathology , Humans , MAP Kinase Kinase Kinases/genetics , Male , Neurons/pathology , Phosphorylation/genetics , Protein Serine-Threonine Kinases , tau Proteins/genetics
4.
J Biol Chem ; 286(34): 30161-70, 2011 Aug 26.
Article in English | MEDLINE | ID: mdl-21705329

ABSTRACT

Prostate-derived sterile 20-like kinases (PSKs) 1-α, 1-ß, and 2 are members of the germinal-center kinase-like sterile 20 family of kinases. Previous work has shown that PSK 1-α binds and stabilizes microtubules whereas PSK2 destabilizes microtubules. Here, we have investigated the activation and autophosphorylation of endogenous PSKs and show that their catalytic activity increases as cells accumulate in G(2)/M and declines as cells exit mitosis. PSKs are stimulated in synchronous HeLa cells as they progress through mitosis, and these proteins are activated catalytically during each stage of mitosis. During prophase and metaphase activated PSKs are located in the cytoplasm and at the spindle poles, and during telophase and cytokinesis stimulated PSKs are present in trans-Golgi compartments. In addition, small interfering RNA (siRNA) knockdown of PSK1-α/ß or PSK2 expression inhibits mitotic cell rounding as well as spindle positioning and centralization. These results show that PSK catalytic activity increases during mitosis and suggest that these proteins can contribute functionally to mitotic cell rounding and spindle centralization during cell division.


Subject(s)
Cell Division/physiology , Microtubules/metabolism , Mitosis/physiology , Protein Kinases/metabolism , Spindle Apparatus/metabolism , trans-Golgi Network/metabolism , Enzyme Activation/physiology , HeLa Cells , Humans , Microtubules/genetics , Protein Kinases/genetics , Spindle Apparatus/genetics , trans-Golgi Network/genetics
5.
J Biol Chem ; 282(9): 6484-93, 2007 Mar 02.
Article in English | MEDLINE | ID: mdl-17158878

ABSTRACT

We have demonstrated previously that full-length prostate-derived sterile 20-like kinase 1-alpha (PSK1-alpha) binds to microtubules via its C terminus and regulates their organization and stability independently of its catalytic activity. Here we have shown that apoptotic and microtubule-disrupting agents promote catalytic activation, C-terminal cleavage, and nuclear translocation of endogenous phosphoserine 181 PSK1-alpha and activated N-terminal PSK1-alpha-induced apoptosis. PSK1-alpha, unlike its novel isoform PSK1-beta, stimulated the c-Jun N-terminal kinase (JNK) pathway, and the nuclear localization of PSK1-alpha and its induction of cell contraction, membrane blebbing, and apoptotic body formation were dependent on JNK activity. PSK1-alpha was also a caspase substrate, and the broad spectrum caspase inhibitor benzyloxycarbonyl-VAD-fluoromethyl ketone or mutation of a putative caspase recognition motif ((916)DPGD(919)) blocked nuclear localization of PSK1-alpha and its induction of membrane blebs. Additional inhibition of caspase 9 was needed to prevent cell contraction. PSK1-alpha is therefore a bifunctional kinase that associates with microtubules, and JNK- and caspase-mediated removal of its C-terminal microtubule-binding domain permits nuclear translocation of the N-terminal region of PSK1-alpha and its induction of apoptosis.


Subject(s)
Active Transport, Cell Nucleus , Apoptosis , Caspases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Protein Kinases/physiology , Animals , Cell Line, Tumor , Cell Membrane , Cell Shape , Humans , Transfection
6.
J Biol Chem ; 281(11): 7317-23, 2006 Mar 17.
Article in English | MEDLINE | ID: mdl-16407310

ABSTRACT

We have reported previously that human prostate-derived sterile 20-like kinase (PSK) 1 alters actin cytoskeletal organization and binds to microtubules, regulating their organization and stability. We have shown a structurally related protein kinase PSK2, which lacks a microtubule-binding site, activated c-Jun N-terminal kinase (JNK), and induced apoptotic morphological changes that include cell contraction, membrane blebbing, and apoptotic body formation. Apoptotic stimuli increased the catalytic activity of endogenous PSK2 and JNK, and dominant negative JNK or a physiological inhibitor of JNK blocked these apoptotic morphological responses to PSK2, demonstrating a requirement for JNK. PSK2 also stimulated the cleavage of Rho kinase-1 (ROCK-I), and the activity of ROCK-I was required for PSK2 to induce cell contraction and membrane blebbing. The activation of caspases was also needed for the induction of membrane blebbing by PSK2, which was itself a substrate for caspase 3. PSK2 therefore regulates apoptotic morphology associated with the execution phase of apoptosis, which involves dynamic reorganization of the actin cytoskeleton, via downstream targets that include JNK and ROCK-I. Our findings suggest that PSKs form a subgroup of sterile 20 (STE20)-like kinases that regulate different cytoskeletal processes.


Subject(s)
Apoptosis , JNK Mitogen-Activated Protein Kinases/metabolism , Protein Kinases/physiology , Protein Serine-Threonine Kinases/metabolism , Actin Depolymerizing Factors/chemistry , Actins/metabolism , Binding Sites , Blotting, Northern , Caspase 2 , Caspase 3 , Caspases/metabolism , Catalysis , Cell Membrane/metabolism , Cytoskeleton/metabolism , DNA Fragmentation , Genes, Dominant , Humans , Immunoblotting , Immunoprecipitation , Intracellular Signaling Peptides and Proteins , MAP Kinase Kinase 4/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Microtubules/metabolism , Models, Genetic , Plasmids/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/chemistry , Sorbitol/pharmacology , Staurosporine/chemistry , Staurosporine/pharmacology , Time Factors , rho-Associated Kinases
7.
J Pharm Pharmacol ; 54(8): 1147-50, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12195832

ABSTRACT

Galangin is a flavanol with several biological activities. We have evaluated the effect of galangin on the contractile response elicited by electrical field stimulation (EFS) in the rat isolated urinary bladder. Galangin (10(-8)-10(-4) M) produced a concentration-dependent inhibition of the EFS contractile response without modifying the contractions produced by exogenous acetylcholine (10(-6) M). Blockade of adrenergic and cholinergic nerves with a combination of atropine (10(-6) M), phentolamine (10(-6) M) and propranolol (10(-6) M) or blockade of tachykinin NK1 and NK2 receptors with SR140333 (10(-7) M) and SR48968 (10(-6) M) did not modify the inhibitory effect of galangin. However, verapamil (10(-7) M) significantly reduced the inhibitory effect of galangin. It is concluded that the galangin inhibits EFS-induced contractions of the rat urinary bladder by acting on L-type calcium channels on presynaptic nerves.


Subject(s)
Calcium Channels/drug effects , Flavonoids/pharmacology , Mutagens/pharmacology , Urinary Bladder/drug effects , Urinary Bladder/physiology , Animals , Calcium Channels/physiology , Electric Stimulation , Male , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth/physiology , Organ Culture Techniques , Presynaptic Terminals/drug effects , Presynaptic Terminals/physiology , Rats , Rats, Wistar , Urinary Bladder/innervation
8.
J Gastroenterol Hepatol ; 13(S3): S190-S192, 1998 Nov.
Article in English | MEDLINE | ID: mdl-28976651

ABSTRACT

Gastrointestinal damage by non-steroidal anti-inflammatory drugs (NSAID), which is common and sometimes fatal, involves inhibition of prostaglandin (PG) synthesis. The damage is less with NSAID that inhibit inflammatory cyclo-oxygenase (COX)-2 but not gastroprotective COX-1. We have compared nimesulide and acemetacin, two NSAID that cause relatively little gastric damage, with indomethacin for effects on purified COX-1 and COX-2. The results are related to findings on human gastric mucosa and leucocytes. With purified COX-1, inhibition was absent with nimesulide, weak with acemetacin (inhibitory concentration of 50% [IC50 ] 85 m̈mol/L), and potent with indomethacin (IC50 0.6 m̈mol/L). Inhibition of purified COX-2 occurred with nimesulide and indomethacin (IC50 values 90 and 4.1 m̈mol/L, respectively) but not acemetacin. All results with nimesulide are consistent with a preferential block of COX-2 that contributes to relatively little gastric damage in patients. However, our previous report on acemetacin needs re-evaluation. Acemetacin did not inhibit sheep COX-2 and only weakly inhibited COX-1. Our previous leucocyte (COX-2) experiments required 24 h incubation, and hydrolysis of acemetacin to indomethacin presumably accounted for the COX-2 inhibition. In the few minutes of enzyme incubation, virtually no hydrolysis would be expected. Acemetacin itself presumably causes relatively little gastric damage mainly because it only weakly inhibits COX-1.

SELECTION OF CITATIONS
SEARCH DETAIL
...