Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Cancer Sci ; 114(11): 4299-4313, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37700438

ABSTRACT

N6-methyladenosine (m6A) is the most abundant internal modification in eukaryotic RNA and involved in the carcinogenesis of various malignancies. However, the functions and mechanisms of m6A in gallbladder cancer (GBC) remain unclear. In this study, we investigated the role and underlying mechanism of the RNA-binding protein YT521-B homology domain-containing family protein 2 (YTHDF2), an m6A reader, in GBC. Herein, we detected that YTHDF2 was remarkably upregulated in GBC tissues compared to normal gallbladder tissues. Functionally, YTHDF2 overexpression promoted the proliferation, tumor growth, migration, and invasion of GBC cells while inhibiting the apoptosis in vitro and in vivo. Conversely, YTHDF2 knockdown induced opposite results. Mechanistically, we further investigated the underlying mechanism by integrating RNA immunoprecipitation sequencing (RIP-seq), m6A-modified RIP-seq, and RNA sequencing, which revealed that death-associated protein kinase 3 (DAPK3) is a direct target of YTHDF2. YTHDF2 binds to the 3'-UTR of DAPK3 mRNA and facilitates its degradation in an m6A-dependent manner. DAPK3 inhibition restores the tumor-suppressive phenotype induced by YTHDF2 deficiency. Moreover, the YTHDF2/DAPK3 axis induces the resistance of GBC cells to gemcitabine. In conclusion, we reveal the oncogenic role of YTHDF2 in GBC, demonstrating that YTHDF2 increases the mRNA degradation of the tumor suppressor DAPK3 in an m6A-dependent way, which promotes GBC progression and desensitizes GBC cells to gemcitabine. Our findings provide novel insights into potential therapeutic strategies for GBC.


Subject(s)
Gallbladder Neoplasms , Gemcitabine , Humans , Gallbladder Neoplasms/drug therapy , Gallbladder Neoplasms/genetics , Gallbladder Neoplasms/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , RNA , Death-Associated Protein Kinases/metabolism
2.
Biochem Biophys Res Commun ; 625: 122-127, 2022 10 15.
Article in English | MEDLINE | ID: mdl-35961135

ABSTRACT

Human immunodeficiency virus-1 (HIV-1) infection leads to the development of acquired immunodeficiency syndrome (AIDS). To establish a productive infection, HIV-1 hijacks the cellular machinery and modulates various physiological processes to propagate itself. The pathways altered by HIV-1 include cell cycle, autophagy, apoptosis, cell stress pathways, immune response, antiviral response, etc. Zipper interacting protein kinase (ZIPK) is a member of the death-associated protein kinase (DAPK) family of proteins, known to be one of the key regulators of cell death and cell survival pathways. ZIPK is also involved in regulating many cellular processes that are altered during HIV-1 infection; thus, we have explored the functional role of ZIPK in HIV-1 infection. Our results show that ZIPK protein expression is downregulated during HIV-1 infection in Nef dependent manner. Overexpression of ZIPK leads to downregulation in LTR-driven gene expression and virus production, whereas ZIPK knockdown induces viral gene expression and replication. HIV-1 promoter activity is reportedly enhanced by Nef-mediated activation of some transcription factors like NFκB and STAT3. ZIPK is reported to inhibit the STAT3 activity by phosphorylating it at ser-727. Our results show that STAT3 (ser-727) phosphorylation is decreased upon overexpression of Nef with simultaneous downregulation of ZIPK expression. We finally show that HIV-1 Nef interacts with ZIPK and induces its proteasomal degradation. Overall, our data suggests that Nef is involved in downregulation of ZIPK thereby increasing the virus production through rescue of STAT3 activity.


Subject(s)
Gene Products, nef , HIV-1 , Death-Associated Protein Kinases , Gene Products, nef/physiology , HIV-1/genetics , Humans , Protein Kinases , Viral Proteins , Virus Replication , nef Gene Products, Human Immunodeficiency Virus/genetics
3.
Biochem Biophys Res Commun ; 533(4): 1095-1101, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33032825

ABSTRACT

Death-associated protein kinase 3 (DAPK3), a member of the DAPK family, contributes to cytokinesis by phosphorylating myosin II regulatory light chain (MRLC). Missense mutations in DAPK3, T112M, D161N, and P216S, were observed in the lung, colon, and cervical cancers, respectively, but the effects of these mutations on cytokinesis remain unclear. Here, we show that cells expressing EGFP-DAPK3-T112M, -D161N, or -P216S exhibited reduced rates of cytokinesis, with an increased ratio of multinucleated cells. In addition, these cells exhibited reduced levels of phosphorylated MRLC at the contractile ring. Collectively, our data demonstrates that cancer-associated DAPK3 mutations impair cytokinesis by reducing phosphorylated MRLC.


Subject(s)
Cytokinesis/genetics , Death-Associated Protein Kinases/genetics , Myosin Light Chains/metabolism , Death-Associated Protein Kinases/metabolism , HeLa Cells , Humans , Mutation, Missense , Phosphorylation
4.
IUBMB Life ; 71(6): 738-748, 2019 06.
Article in English | MEDLINE | ID: mdl-30548122

ABSTRACT

For many decades, kinases have predominantly been characterized as oncogenes and drivers of tumorigenesis, because activating mutations in kinases occur in cancer with high frequency. The oncogenic functions of kinases relate to their roles as growth factor receptors and as critical mediators of mitogen-activated pathways. Indeed, some of the most promising cancer therapeutic agents are kinase inhibitors. However, cancer genomics studies, especially screens that utilize high-throughput identification of loss-of-function somatic mutations, are beginning to shed light on a widespread role for kinases as tumor suppressors. The initial characterization of tumor-suppressing kinases- in particular members of the protein kinase C (PKC) family, MKK4 of the mitogen-activated protein kinase kinase family, and DAPK3 of the death-associated protein kinase family- laid the foundation for bioinformatic approaches that enable the identification of other tumor-suppressing kinases. In this review, we discuss the important role that kinases play as tumor suppressors, using several examples to illustrate the history of their discovery and highlight the modern approaches that presently aid in the identification of tumor-suppressing kinases. © 2018 IUBMB Life, 71(6):738-748, 2019.


Subject(s)
Death-Associated Protein Kinases/genetics , MAP Kinase Kinase 4/genetics , Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Carcinogenesis/genetics , Humans , Protein Kinase C/genetics , Signal Transduction , Tumor Suppressor Proteins/chemistry
5.
Proteins ; 86(11): 1211-1217, 2018 11.
Article in English | MEDLINE | ID: mdl-30381843

ABSTRACT

Zipper-interacting protein kinase (ZIPK) is a Ser/Thr kinase that mediates a variety of cellular functions. Analogue-sensitive kinase technology was applied to the study of ZIPK signaling in coronary artery smooth muscle cells. ZIPK was engineered in the ATP-binding pocket by substitution of a bulky gatekeeper amino acid (Leu93) with glycine. Cell-permeable derivatives of pyrazolo[3,4-d]pyrimidine provided effective inhibition of L93G-ZIPK (1NM-PP1, IC50 , 1.0 µM; 3MB-PP1, IC50 , 2.0 µM; and 1NA-PP1, IC50 , 8.6 µM) but only 3MB-PP1 had inhibitory potential (IC50 > 10 µM) toward wild-type ZIPK. Each of the compounds also attenuated Rho-associated coiled-coil containing protein kinase (ROCK) activity under experimental conditions found to be optimal for inhibition of L93G-ZIPK. In silico molecular simulations showed effective docking of 1NM-PP1 into ZIPK following mutational enlargement of the ATP-binding pocket. Molecular simulation of 1NM-PP1 docking in the ATP-binding pocket of ROCK was also completed. The 1NM-PP1 inhibitor was selected as the optimal compound for selective chemical genetics in smooth muscle cells since it displayed the highest potency for L93G-ZIPK relative to WT-ZIPK and the weakest off-target effects against other relevant kinases. Finally, the 1NM-PP1 and L93G-ZIPK pairing was effectively applied in vascular smooth muscle cells to manipulate the phosphorylation level of LC20, a previously defined target of ZIPK.


Subject(s)
Adenosine Triphosphate/metabolism , Death-Associated Protein Kinases/metabolism , Signal Transduction , Binding Sites/drug effects , Cell Line , Coronary Vessels/cytology , Coronary Vessels/metabolism , Death-Associated Protein Kinases/antagonists & inhibitors , Death-Associated Protein Kinases/chemistry , Death-Associated Protein Kinases/genetics , Humans , Molecular Docking Simulation , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Protein Engineering , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Transfection
6.
PNAS Nexus ; 3(9): pgae401, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39319326

ABSTRACT

Sixteen patient-derived xenografts (PDXs) were analyzed using a mass spectrometry (MS)-based kinase inhibitor pull-down assay (KIPA), leading to the observation that death-associated protein kinase 3 (DAPK3) is significantly and specifically overexpressed in the triple-negative breast cancer (TNBC) models. Validation studies confirmed enrichment of DAPK3 protein, in both TNBC cell lines and tumors, independent of mRNA levels. Genomic knockout of DAPK3 in TNBC cell lines inhibited in vitro migration and invasion, along with down-regulation of an epithelial-mesenchymal transition (EMT) signature, which was confirmed in vivo. The kinase and leucine-zipper domains within DAPK3 were shown by a mutational analysis to be essential for functionality. Notably, DAPK3 was found to inhibit the levels of desmoplakin (DSP), a crucial component of the desmosome complex, thereby explaining the observed migration and invasion effects. Further exploration with immunoprecipitation-mass spectrometry (IP-MS) identified that leucine-zipper protein 1 (LUZP1) is a preferential binding partner of DAPK3. LUZP1 engages in a leucine-zipper domain-mediated interaction that protects DAPK3 from proteasomal degradation. Thus, the DAPK3/LUZP1 heterodimer emerges as a newly discovered regulator of EMT/desmosome components that promote TNBC cell migration.

7.
Article in English | MEDLINE | ID: mdl-38563090

ABSTRACT

In the brain, environmental changes, such as neuroinflammation, can induce senescence, characterized by the decreased proliferation of neurons and dendrites and synaptic and vascular damage, resulting in cognitive decline. Senescence promotes neuroinflammatory disorders by senescence-associated secretory phenotypes and reactive oxygen species. In human brain microvascular endothelial cells (HBMVECs), we demonstrate that chronological aging and irradiation increase death-associated protein kinase 3 (DAPK3) expression. To confirm the role of DAPK3 in HBMVEC senescence, we disrupted DAPK3 activity using small interfering RNA (siRNA) or a dominant-negative mutant (DAPK3-P216S), which reduced cellular senescence phenotypes, as assessed by changes in tube formation, senescence-associated beta-galactosidase activity, and cell proliferation. In endothelial cells, DAPK3 promotes cellular senescence by regulating the phosphorylation and inactivation of peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC1α) via the protein kinase B pathway, resulting in the decreased expression of mitochondrial metabolism-associated genes, such as ATP5G1, BDNF, and COX5A. Our studies show that DAPK3 is involved in cellular senescence and PGC1α regulation, suggesting that DAPK3 regulation may be important for treating aging-related brain diseases or the response to radiation therapy.


Subject(s)
Cellular Senescence , Endothelial Cells , Humans , Endothelial Cells/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Cellular Senescence/physiology , Cell Proliferation/genetics , Brain/metabolism , RNA, Small Interfering/metabolism , Death-Associated Protein Kinases/genetics , Death-Associated Protein Kinases/metabolism
8.
FEBS Lett ; 597(5): 643-656, 2023 03.
Article in English | MEDLINE | ID: mdl-36723402

ABSTRACT

Stiffness of the extracellular matrix regulates various biological responses, but the response mechanisms are poorly understood. Here, we found that the nuclear diphosphorylated myosin regulatory light chain (2P-MRLC) is a critical mechanomediator that suppresses apoptosis in response to substrate stiffness. Stiff substrates promoted the nuclear localization of 2P-MRLC. Zipper-interacting protein kinase [ZIPK; also known as death-associated protein kinase 3 (DAPK3)], a kinase for MRLC, was localized in the nucleus in response to stiff substrates and promoted the nuclear localization of 2P-MRLC. Moreover, actin fiber formation induced by substrate stiffness promoted the nuclear localization of 2P-MRLC via ZIPK. 2P-MRLC in response to substrate stiffness suppressed the expression of MAF bZIP transcription factor B (MafB) and repressed apoptosis. These findings reveal a newly identified role of MRLC in mechanotransduction.


Subject(s)
Mechanotransduction, Cellular , Myosin Light Chains , Myosin Light Chains/metabolism , Phosphorylation , Actins/metabolism , Apoptosis
9.
Chin J Integr Med ; 27(12): 896-904, 2021 Dec.
Article in English | MEDLINE | ID: mdl-31418133

ABSTRACT

OBJECTIVE: To investigate a Met-controlled allosteric module (AM) of neural generation as a potential therapeutic target for brain ischemia. METHODS: We selected Markov clustering algorithm (MCL) to mine functional modules in the related target networks. According to the topological similarity, one functional module was predicted in the modules of baicalin (BA), jasminoidin (JA), cholic acid (CA), compared with I/R model modules. This functional module included three genes: Inppl1, Met and Dapk3 (IMD). By gene ontology enrichment analysis, biological process related to this functional module was obtained. This functional module participated in generation of neurons. Western blotting was applied to present the compound-dependent regulation of IMD. Co-immunoprecipitation was used to reveal the relationship among the three members. We used IF to determine the number of newborn neurons between compound treatment group and ischemia/reperfusion group. The expressions of vascular endothelial growth factor (VEGF) and matrix metalloproteinase 9 (MMP-9) were supposed to show the changing circumstances for neural generation under cerebral ischemia. RESULTS: Significant reduction in infarction volume and pathological changes were shown in the compound treatment groups compared with the I/R model group (P<0.05). Three nodes in one novel module of IMD were found to exert diverse compound-dependent ischemic-specific excitatory regulatory activities. An anti-ischemic excitatory allosteric module (AME) of generation of neurons (AME-GN) was validated successfully in vivo. Newborn neurons increased in BJC treatment group (P<0.05). The expression of VEGF and MMP-9 decreased in the compound treatment groups compared with the I/R model group (P<0.05). CONCLUSIONS: AME demonstrates effectiveness of our pioneering approach to the discovery of therapeutic target. The novel approach for AM discovery in an effort to identify therapeutic targets holds the promise of accelerating elucidation of underlying pharmacological mechanisms in cerebral ischemia.


Subject(s)
Brain Ischemia , Gene Regulatory Networks , Proto-Oncogene Proteins c-met , Algorithms , Animals , Brain Ischemia/drug therapy , Gene Ontology , Markov Chains , Matrix Metalloproteinase 9 , Rodentia , Vascular Endothelial Growth Factor A
10.
Mol Cell Endocrinol ; 502: 110674, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31811899

ABSTRACT

Autophagy plays an essential role in gestational diabetes mellitus (GDM). Death-associated protein kinase-3 (DAPK3) regulates a variety of cellular functions; however, the relationship between DAPK3 and autophagy is unknown. In this study, we aim to investigate whether DAPK3 is associated with autophagy in GDM, and we found that DAPK3 was upregulated in the placenta of GDM patients and extravillous trophoblast cells under high-glucose conditions. Silencing DAPK3 decreased the assembly of the STX17-SNAP29-VAMP8 complex, leading to the blockade of autophagosome-lysosome fusion by mediating synaptosomal-associated protein 29 (SNAP29). Moreover, knockdown of DAPK3 ameliorates cell invasion and mediates autophagy in high glucose, and does not alter the expression of autophagy-related genes in normal glucose. Our study demonstrates the significance of DAPK3 in autophagy and GDM, which may provide new insights into the molecular mechanisms regulating trophoblast invasion.


Subject(s)
Autophagosomes/metabolism , Death-Associated Protein Kinases/genetics , Diabetes, Gestational/genetics , Lysosomes/metabolism , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/metabolism , Cell Line , Cell Movement , Death-Associated Protein Kinases/metabolism , Diabetes, Gestational/metabolism , Female , Gene Knockdown Techniques , Glucose/adverse effects , Humans , Pregnancy , Trophoblasts/cytology , Trophoblasts/drug effects , Trophoblasts/metabolism , Up-Regulation
11.
Mol Oncol ; 14(6): 1268-1281, 2020 06.
Article in English | MEDLINE | ID: mdl-32306542

ABSTRACT

Cross-linking of the B-cell receptor (BCR) induces transcriptional activation of immediate early genes (IEGs) including EGR1 and DUSP2 in chronic lymphocytic leukaemia (CLL). Here, we have shown that this transcriptional activation correlated with histone H3 threonine 6 and 11 phosphorylation. Both transcription and histone post-translational modifications are repressed by ibrutinib, a small molecule inhibitor used in CLL treatment. Moreover, we have identified the death-associated protein kinase 3 (DAPK3), as the kinase mediating these histone phosphorylation marks in response to activation of the BCR signalling pathway with this kinase being recruited to RNA polymerase II in an anti-IgM-dependent manner. DAPK inhibition mimics ibrutinib-induced repression of both IEG mRNA and histone H3 phosphorylation and has anti-proliferative effect comparable to ibrutinib in CLL in vitro. DAPK inhibitor does not repress transcription itself but impacts on mRNA processing and has a broader anti-tumour effect than ibrutinib, by repressing both anti-IgM- and CD40L-dependent activation.


Subject(s)
Death-Associated Protein Kinases/genetics , Genes, Immediate-Early , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , RNA Processing, Post-Transcriptional/genetics , CD40 Ligand/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Death-Associated Protein Kinases/antagonists & inhibitors , Death-Associated Protein Kinases/metabolism , Genetic Loci , Histones/metabolism , Humans , Immunoglobulin M/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Phosphorylation/drug effects , Phosphothreonine/metabolism , Protein Kinase Inhibitors/pharmacology , RNA Processing, Post-Transcriptional/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Antigen, B-Cell/metabolism
12.
Eur J Pharmacol ; 852: 90-98, 2019 Jun 05.
Article in English | MEDLINE | ID: mdl-30851272

ABSTRACT

Vascular calcification (VC) is a critical feature of chronic kidney disease (CKD), diabetes, hypertension, and atherosclerosis. Death-associated protein kinase 3 (DAPK3) is involved in vascular remodeling in hypertension. However, it remains to be clarified whether DAPK3 controls vascular smooth muscle cell (VSMC) phenotypic transition into an osteogenic cell phenotype, which is an important process for VC. In vivo VC was induced in rats by vitamin D3 and nicotine. VSMCs were incubated with calcifying media containing ß-glycerophosphate and Ca2+ to induce VC in vitro. Herein, we demonstrated increased expression of DAPK3 in the aortas of VC rats and VSMCs cultured in calcifying media. Knockdown of DAPK3 significantly inhibited calcifying media-induced VSMC mineralization and retarded the phenotypic transformation of VSMCs into osteogenic cells. Silencing of DAPK3 suppressed endoplasmic reticulum stress (ERS) related protein expressions, but upregulated the phosphorylation level of AMP-activated protein kinase (AMPK) in calcified VSMCs. Moreover, pretreatment with AMPK inhibitor Compound C abolished DAPK3 shRNA-mediated inhibition of ERS in VSMCs. In vivo, DAPK inhibitor significantly prevented calcium deposition in the aortas of VC rats. The present results revealed that DAPK3 modulated VSMC calcification through AMPK-mediated ERS signaling.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Death-Associated Protein Kinases/deficiency , Death-Associated Protein Kinases/genetics , Endoplasmic Reticulum Stress/genetics , Gene Knockdown Techniques , Vascular Calcification/pathology , Animals , Death-Associated Protein Kinases/antagonists & inhibitors , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Male , Muscle, Smooth, Vascular/metabolism , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Vascular Calcification/genetics , Vascular Calcification/metabolism
13.
Methods Mol Biol ; 1923: 187-209, 2019.
Article in English | MEDLINE | ID: mdl-30737741

ABSTRACT

New mass spectrometry approaches enable antibody-independent tracking of protein production. Herein, we outline an antibody-independent mass spectrometry method for tracking recombinant protein production in the methylotrophic yeast Pichia pastoris system.


Subject(s)
Mass Spectrometry/methods , Pichia/metabolism , Proteomics/methods , Recombinant Proteins/metabolism , Pichia/genetics , Recombinant Proteins/genetics
14.
Cell Chem Biol ; 25(10): 1195-1207.e32, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30033129

ABSTRACT

Sustained vascular smooth muscle hypercontractility promotes hypertension and cardiovascular disease. The etiology of hypercontractility is not completely understood. New therapeutic targets remain vitally important for drug discovery. Here we report that Pim kinases, in combination with DAPK3, regulate contractility and control hypertension. Using a co-crystal structure of lead molecule (HS38) in complex with DAPK3, a dual Pim/DAPK3 inhibitor (HS56) and selective DAPK3 inhibitors (HS94 and HS148) were developed to provide mechanistic insight into the polypharmacology of hypertension. In vitro and ex vivo studies indicated that Pim kinases directly phosphorylate smooth muscle targets and that Pim/DAPK3 inhibition, unlike selective DAPK3 inhibition, significantly reduces contractility. In vivo, HS56 decreased blood pressure in spontaneously hypertensive mice in a dose-dependent manner without affecting heart rate. These findings suggest including Pim kinase inhibition within a multi-target engagement strategy for hypertension management. HS56 represents a significant step in the development of molecularly targeted antihypertensive medications.


Subject(s)
Death-Associated Protein Kinases/antagonists & inhibitors , Hypertension/drug therapy , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Amino Acid Sequence , Animals , Blood Pressure/drug effects , Crystallography, X-Ray , Death-Associated Protein Kinases/chemistry , Death-Associated Protein Kinases/metabolism , Humans , Hypertension/metabolism , Hypertension/physiopathology , Male , Mice , Models, Molecular , Molecular Targeted Therapy , Muscle Contraction/drug effects , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/chemistry , Proto-Oncogene Proteins c-pim-1/metabolism , Rats, Sprague-Dawley , Sequence Alignment
15.
Article in English | WPRIM | ID: wpr-922097

ABSTRACT

OBJECTIVE@#To investigate a Met-controlled allosteric module (AM) of neural generation as a potential therapeutic target for brain ischemia.@*METHODS@#We selected Markov clustering algorithm (MCL) to mine functional modules in the related target networks. According to the topological similarity, one functional module was predicted in the modules of baicalin (BA), jasminoidin (JA), cholic acid (CA), compared with I/R model modules. This functional module included three genes: Inppl1, Met and Dapk3 (IMD). By gene ontology enrichment analysis, biological process related to this functional module was obtained. This functional module participated in generation of neurons. Western blotting was applied to present the compound-dependent regulation of IMD. Co-immunoprecipitation was used to reveal the relationship among the three members. We used IF to determine the number of newborn neurons between compound treatment group and ischemia/reperfusion group. The expressions of vascular endothelial growth factor (VEGF) and matrix metalloproteinase 9 (MMP-9) were supposed to show the changing circumstances for neural generation under cerebral ischemia.@*RESULTS@#Significant reduction in infarction volume and pathological changes were shown in the compound treatment groups compared with the I/R model group (P<0.05). Three nodes in one novel module of IMD were found to exert diverse compound-dependent ischemic-specific excitatory regulatory activities. An anti-ischemic excitatory allosteric module (AM@*CONCLUSIONS@#AM


Subject(s)
Animals , Brain Ischemia/drug therapy , Gene Ontology , Gene Regulatory Networks , Rodentia , Vascular Endothelial Growth Factor A
16.
Gene ; 580(1): 17-25, 2016 Apr 10.
Article in English | MEDLINE | ID: mdl-26748242

ABSTRACT

DAPK3 belongs to family of DAPK (death-associated protein kinases) and is involved in the regulation of progression of the cell cycle, cell proliferation, apoptosis and autophagy. It is considered as a tumor suppressor kinase, suggesting the loss of its function in case of certain specific mutations. The T112M, D161N and P216S mutations in DAPK3 have been observed in cancer patients. These DAPK3 mutants have been associated with very low kinase activity, which results in the cellular progression towards cancer. However, a clear understanding of the structural and biophysical variations that occur in DAPK3 with these mutations, resulting in the decreased kinase activity has yet not been deciphered. We performed a molecular dynamic simulation study to investigate such structural variations. Our results revealed that mutations caused a significant structural variation in DAPK3, majorly concentrated in the flexible loops that form part of the ATP binding pocket. Interestingly, D161N and P216S mutations collapsed the ATP binding pocket through flexible loops invasion, hindering ATP binding which resulted in very low kinase activity. On the contrary, T112M mutant DAPK3 reduces ATP binding potential through outward distortion of flexible loops. In addition, the mutant lacked characteristic features of the active protein kinase including proper interaction between HR/FD and DFG motifs, well structured hydrophobic spine and Lys42-Glu64 salt bridge interaction. These observations could possibly explain the underlying mechanism associated with the loss of kinase activity with T112M, D161N and P216S mutation in DAPK3.


Subject(s)
Adenosine Triphosphate/metabolism , Binding Sites/physiology , Death-Associated Protein Kinases/genetics , Molecular Dynamics Simulation , Neoplasms/genetics , Apoptosis/genetics , Autophagy/genetics , Cell Cycle/genetics , Cell Proliferation/genetics , Crystallography, X-Ray , Death-Associated Protein Kinases/ultrastructure , Humans , Models, Molecular , Mutation , Phosphorylation , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL