Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Nature ; 508(7497): 541-5, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24670654

ABSTRACT

Akt, also known as protein kinase B, plays key roles in cell proliferation, survival and metabolism. Akt hyperactivation contributes to many pathophysiological conditions, including human cancers, and is closely associated with poor prognosis and chemo- or radiotherapeutic resistance. Phosphorylation of Akt at S473 (ref. 5) and T308 (ref. 6) activates Akt. However, it remains unclear whether further mechanisms account for full Akt activation, and whether Akt hyperactivation is linked to misregulated cell cycle progression, another cancer hallmark. Here we report that Akt activity fluctuates across the cell cycle, mirroring cyclin A expression. Mechanistically, phosphorylation of S477 and T479 at the Akt extreme carboxy terminus by cyclin-dependent kinase 2 (Cdk2)/cyclin A or mTORC2, under distinct physiological conditions, promotes Akt activation through facilitating, or functionally compensating for, S473 phosphorylation. Furthermore, deletion of the cyclin A2 allele in the mouse olfactory bulb leads to reduced S477/T479 phosphorylation and elevated cellular apoptosis. Notably, cyclin A2-deletion-induced cellular apoptosis in mouse embryonic stem cells is partly rescued by S477D/T479E-Akt1, supporting a physiological role for cyclin A2 in governing Akt activation. Together, the results of our study show Akt S477/T479 phosphorylation to be an essential layer of the Akt activation mechanism to regulate its physiological functions, thereby providing a new mechanistic link between aberrant cell cycle progression and Akt hyperactivation in cancer.


Subject(s)
Cell Cycle/physiology , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/genetics , Cell Proliferation , Cyclin A2/metabolism , Cyclin-Dependent Kinase 2/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Enzyme Activation , Male , Mechanistic Target of Rapamycin Complex 2 , Mice , Multiprotein Complexes/metabolism , Neoplasms/enzymology , Neoplasms/pathology , Olfactory Bulb/cytology , Olfactory Bulb/enzymology , Olfactory Bulb/metabolism , Oncogene Protein v-akt/chemistry , Oncogene Protein v-akt/metabolism , Phosphorylation , Phosphoserine/metabolism , Phosphothreonine/metabolism , TOR Serine-Threonine Kinases/metabolism
2.
Int J Mol Sci ; 20(5)2019 Mar 03.
Article in English | MEDLINE | ID: mdl-30832444

ABSTRACT

Several reports have shown that thymoquinone (TQ) effectively attenuates angiogenesis in cancer cells, resulting in suppression of tumor growth. However, it is not yet clear whether TQ reduces hypoxia-inducible factor-1α (HIF-1α) expression in hypoxic cancer cells. Here, we found that TQ was a novel HIF-1α inhibitor through hypoxia response element (HRE)-luciferase assay-based large screening by using 502 natural compounds containing chemical library. TQ reduced HIF-1α protein levels in renal cancer cells; however, it did not affect the HIF-1α protein levels in the presence of proteasome inhibitor, MG132, indicating that the reduction effects of TQ on HIF-1α protein are mediated via the ubiquitination-proteasome dependent pathway. TQ boosted HIF-1α protein degradation, and the mechanism was revealed by inhibiting interaction between HSP90 and HIF-1α. TQ suppressed downstream genes of HIF-1α, indicating negative impact of TQ on HIF-1α transcriptional activities. In addition, TQ altered glucose, lactate, and ATP levels, leading to anaerobic metabolic disturbance. TQ induced apoptosis in hypoxic cancer cells as determined by crystal violet staining and flow cytometry for annexin V-stained cells. Taken together, we suggested that TQ is a potential anticancer agent targeting HIF-1α.


Subject(s)
Antineoplastic Agents/pharmacology , Benzoquinones/pharmacology , Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Apoptosis/drug effects , Cell Hypoxia , Cell Line, Tumor , Humans , Kidney Neoplasms/metabolism
3.
Hum Mol Genet ; 25(12): 2498-2513, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27094130

ABSTRACT

Extracellular deposition of amyloid-beta (Aß) peptide, a metabolite of sequential cleavage of amyloid precursor protein (APP), is a critical step in the pathogenesis of Alzheimer's disease (AD). While death-associated protein kinase 1 (DAPK1) is highly expressed in AD brains and its genetic variants are linked to AD risk, little is known about the impact of DAPK1 on APP metabolism and Aß generation. In this study, we demonstrated a novel effect of DAPK1 in the regulation of APP processing using cell culture and mouse models. DAPK1, but not its kinase deficient mutant (K42A), significantly increased human Aß secretion in neuronal cell culture models. Moreover, knockdown of DAPK1 expression or inhibition of DAPK1 catalytic activity significantly decreased Aß secretion. Furthermore, DAPK1, but not K42A, triggered Thr668 phosphorylation of APP, which may initiate and facilitate amyloidogenic APP processing leading to the generation of Aß. In Tg2576 APPswe-overexpressing mice, knockout of DAPK1 shifted APP processing toward non-amyloidogenic pathway and decreased Aß generation. Finally, in AD brains, elevated DAPK1 levels showed co-relation with the increase of APP phosphorylation. Combined together, these results suggest that DAPK1 promotes the phosphorylation and amyloidogenic processing of APP, and that may serve a potential therapeutic target for AD.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Death-Associated Protein Kinases/genetics , Neurons/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/biosynthesis , Animals , Brain/metabolism , Brain/pathology , Death-Associated Protein Kinases/antagonists & inhibitors , Disease Models, Animal , Gene Expression Regulation , Humans , Mice , Neurons/pathology , Phosphorylation
4.
Int J Mol Sci ; 19(11)2018 Nov 01.
Article in English | MEDLINE | ID: mdl-30388809

ABSTRACT

YAP/TAZ, a transcriptional co-activator of Hippo pathway, has emerged as a central player in vessel homeostasis such as sprouting angiogenesis and vascular barrier stabilization, during development. However, the role of YAP/TAZ in pathological angiogenesis remains unclear. Here, we demonstrated that YAP/TAZ is a critical mediator in leukocyte-endothelial adhesion induced by the vascular inflammatory cytokine TNF-α. YAP/TAZ was dephosphorylated, translocated from the cytosol to the nucleus, and activated by TNF-α in endothelial cells. A specific inhibitor of Rho GTPases suppressed the TNF-α-induced dephosphorylation of YAP. Knockdown of YAP/TAZ using siRNA significantly reduced the expression of the leukocyte adhesion molecule VCAM1 induced by TNF-α. The adhesion of monocytes to endothelial cells was also markedly reduced by YAP/TAZ silencing. However, knockdown of YAP/TAZ did not affect TNF-α-induced NF-κB signaling. Overall, these results suggest that YAP/TAZ plays critical roles in regulating TNF-α-induced endothelial cell adhesive properties without affecting the NF-κB pathway, and implicate YAP/TAZ as a potential therapeutic target for treating inflammatory vascular diseases.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Leukocytes/cytology , Phosphoproteins/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/metabolism , Cell Adhesion/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Intercellular Adhesion Molecule-1/metabolism , Leukocytes/drug effects , Leukocytes/metabolism , NF-kappa B/metabolism , Phosphorylation , Signal Transduction , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins , rho GTP-Binding Proteins/metabolism
5.
Int J Mol Sci ; 19(10)2018 Oct 12.
Article in English | MEDLINE | ID: mdl-30321984

ABSTRACT

Reduced therapeutic efficacy of sorafenib, a first-generation multikinase inhibitor, is often observed during the treatment of advanced hepatocellular carcinoma (HCC). Emodin is an active component of Chinese herbs, and is effective against leukemia, lung cancer, colon cancer, pancreatic cancer, and HCC; however, the sensitizing effect of emodin on sorafenib-based HCC therapy has not been evaluated. Here, we demonstrate that emodin significantly improved the anti-cancer effect of sorafenib in HCC cells, such as HepG2, Hep3B, Huh7, SK-HEP-1, and PLC/PRF5. Mechanistically, emodin inhibits sterol regulatory element-binding protein-2 (SREBP-2) transcriptional activity, which suppresses cholesterol biosynthesis and oncogenic protein kinase B (AKT) signaling. Additionally, attenuated cholesterol synthesis and oncogenic AKT signaling inactivated signal transducer and activator of transcription 3 (STAT3), an oncogenic transcription factor. Furthermore, emodin synergistically increased cell cycle arrest in the G1 phase and apoptotic cells in the presence of sorafenib. Animal models xenografted with HepG2 or SK-HEP-1 cells also showed that the combination of emodin and sorafenib was sufficient to inhibit tumor growth. Overall, these results suggested that the combination of emodin and sorafenib may offer a potential therapy for patients with advanced HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Cholesterol/metabolism , Emodin/administration & dosage , Liver Neoplasms/drug therapy , Sorafenib/administration & dosage , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Emodin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/drug effects , Sorafenib/pharmacology , Sterol Regulatory Element Binding Protein 2/genetics , Xenograft Model Antitumor Assays
6.
Int J Mol Sci ; 18(3)2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28257048

ABSTRACT

Recent studies have shown that vanillin has anti-cancer, anti-mutagenic, and anti-metastatic activity; however, the precise molecular mechanism whereby vanillin inhibits metastasis and cancer progression is not fully elucidated. In this study, we examined whether vanillin has anti-cancer and anti-metastatic activities via inhibition of hypoxia-inducible factor-1α (HIF-1α) in A2058 and A375 human malignant melanoma cells. Immunoblotting and quantitative real time (RT)-PCR analysis revealed that vanillin down-regulates HIF-1α protein accumulation and the transcripts of HIF-1α target genes related to cancer metastasis including fibronectin 1 (FN1), lysyl oxidase-like 2 (LOXL2), and urokinase plasminogen activator receptor (uPAR). It was also found that vanillin significantly suppresses HIF-1α mRNA expression and de novo HIF-1α protein synthesis. To understand the suppressive mechanism of vanillin on HIF-1α expression, chromatin immunoprecipitation was performed. Consequently, it was found that vanillin causes inhibition of promoter occupancy by signal transducer and activator of transcription 3 (STAT3), but not nuclear factor-κB (NF-κB), on HIF1A. Furthermore, an in vitro migration assay revealed that the motility of melanoma cells stimulated by hypoxia was attenuated by vanillin treatment. In conclusion, we demonstrate that vanillin might be a potential anti-metastatic agent that suppresses metastatic gene expression and migration activity under hypoxia via the STAT3-HIF-1α signaling pathway.


Subject(s)
Benzaldehydes/pharmacology , Cell Movement/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Melanoma/genetics , Melanoma/metabolism , STAT3 Transcription Factor/metabolism , Cell Line, Tumor , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptional Activation
7.
Molecules ; 23(1)2017 Dec 24.
Article in English | MEDLINE | ID: mdl-29295560

ABSTRACT

Fascaplysin, a natural product isolated from marine sponges, is a potential candidate for the development of anti-cancer drugs. However, the mechanism underlying its therapeutic effect of strengthening anti-cancer efficacy of other drugs is poorly understood. Here, we found that fascaplysin increases phosphorylation of protein kinase B (PKB), also known as AKT, and adenosine monophosphate-activated protein kinase (AMPK), which are considered therapeutic targets for cancer treatment due to their anti-apoptotic or pro-survival functions in cancer. A cell viability assay revealed that pharmacological suppression of AKT using LY294002 enhanced the anti-cancer effect of fascaplysin in various cancer cells. Similarly, fascaplysin was observed to have improved anti-cancer effects in combination with compound C, a selective AMPK inhibitor. Another challenge showed that fascaplysin increased the efficacy of methotrexate (MTX)-mediated cancer therapy by suppressing genes related to folate and purine metabolism. Overall, these results suggest that fascaplysin may be useful for improving the anti-cancer efficacy of targeted anti-cancer drugs, such as inhibitors of phosphoinositide 3-kinase AKT signaling, and chemotherapeutic agents, such as MTX.


Subject(s)
Adenylate Kinase/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Indoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Adenylate Kinase/metabolism , Cell Line, Tumor , DNA Damage , Drug Synergism , Enzyme Activation/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Methotrexate/pharmacology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Stress, Physiological/drug effects
8.
J Neurochem ; 136(6): 1232-1245, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26710244

ABSTRACT

Alzheimer's disease (AD) is a chronic neurodegenerative disease and the risk of developing it increases with advancing age. In this study, we investigated the protective effects of saikosaponin C (SSc), one of the main bioactive components produced by the traditional Chinese herb, radix bupleuri, the root of Bupleurum falcatum, against AD in various neuronal models. Interestingly, we found that SSc has dual effects on AD by targeting amyloid beta (Aß) and tau, two key proteins in AD. SSc significantly suppressed the release of both Aß peptides 1-40 and 1-42 into cell culture supernatants, though it does not affect BACE1 activity and expression. SSc also inhibited abnormal tau phosphorylation at multiple AD-related residues. Moreover, SSc seems to have beneficial effects on cellular tau function; it accelerated nerve growth factor-mediated neurite outgrowth and increased the assembly of microtubules. In addition, SSc increased synaptic marker proteins such as synaptophysin and PSD-95. Considering its various biological activities, our results suggest that SSc might be a novel therapeutic tool for treating human AD and other neurodegenerative diseases. Tau and amyloid beta are two key features in Alzheimer's disease. Saikosaponin C, an active component of Bupleuri Radix, inhibits abnormal tau phosphorylation and amyloid beta production, thereby promoting synaptic integrity. Saikosaponin C also prevents amyloid beta-induced apoptosis in brain vascular endothelial cells. Therefore, Saikosaponin C may provide a new therapeutic strategy for treatment of neurodegenerative diseases, including Alzheimer's disease.

9.
Biochem Biophys Res Commun ; 470(1): 181-186, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26768359

ABSTRACT

USP7 is a deubiquitinating enzyme that involves the ubiquitin proteasome system (UPS) to maintain regulation of protein synthesis and degradation. The well-known substrate of USP7 is the Mdm2-p53 complex. In fact, several studies have reported that functional inhibition of USP7 induces cancer cell apoptosis through activation of p53. However, the contribution of oxidative or endoplasmic reticulum (ER) stress, which is commonly induced by inhibition of the UPS for USP7 inhibitor-mediated apoptosis in cancer cells, has not been investigated. In contrast to previous reports, we show that p53 is not critical during USP7 inhibitor-induced apoptosis in several cancer cells. Inhibition of deubiquitinating enzyme activities by USP7 inhibitors causes ER stress by accumulating polyubiquitinated proteins in cancer cells. Furthermore, we demonstrate that USP7 inhibitors increase intracellular reactive oxygen species and mainly cause cancer cell apoptosis. Taken together, our results reveal that oxidative and ER stress, rather than the Mdm2-p53 axis, mainly contributes to USP7 inhibitor-mediated apoptosis in cancer cells.


Subject(s)
Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Neoplasms, Experimental/metabolism , Oxidative Stress/drug effects , Ubiquitin Thiolesterase/metabolism , Cell Line, Tumor , Humans , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Reactive Oxygen Species/metabolism , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin-Specific Peptidase 7 , Ubiquitination/drug effects
10.
Neurobiol Dis ; 76: 13-23, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25576397

ABSTRACT

The unique proline isomerase Pin1 is pivotal for protecting against age-dependent neurodegeneration in Alzheimer's disease (AD), with its inhibition providing a molecular link between tangle and plaque pathologies. Pin1 is oxidatively modified in human AD brains, but little is known about its regulatory mechanisms and pathological significance of such Pin1 modification. In this paper, our determination of crystal structures of oxidized Pin1 reveals a series of Pin1 oxidative modifications on Cys113 in a sequential fashion. Cys113 oxidization is further confirmed by generating antibodies specifically recognizing oxidized Cys113 of Pin1. Furthermore, Pin1 oxidation on Cys113 inactivates its catalytic activity in vitro, and Ala point substitution of Cys113 inactivates the ability of Pin1 to isomerize tau as well as to promote protein turnover of tau and APP. Moreover, redox regulation affects Pin1 subcellular localization and Pin1-mediated neuronal survival in response to hypoxia treatment. Importantly, Cys113-oxidized Pin1 is significantly increased in human AD brain comparing to age-matched controls. These results not only identify a novel Pin1 oxidation site to be the critical catalytic residue Cys113, but also provide a novel oxidative regulation mechanism for inhibiting Pin1 activity in AD. These results suggest that preventing Pin1 oxidization might help to reduce the risk of AD.


Subject(s)
Alzheimer Disease/metabolism , Peptidylprolyl Isomerase/chemistry , Peptidylprolyl Isomerase/metabolism , Amyloid beta-Protein Precursor/metabolism , Antibodies , Catalytic Domain , Cell Line, Tumor , Hippocampus/metabolism , Humans , NIMA-Interacting Peptidylprolyl Isomerase , Oxidation-Reduction , Peptidylprolyl Isomerase/immunology , tau Proteins/metabolism
11.
Int J Mol Sci ; 16(11): 26880-913, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26569225

ABSTRACT

Ionizing radiation (IR), such as X-rays and gamma (γ)-rays, mediates various forms of cancer cell death such as apoptosis, necrosis, autophagy, mitotic catastrophe, and senescence. Among them, apoptosis and mitotic catastrophe are the main mechanisms of IR action. DNA damage and genomic instability contribute to IR-induced cancer cell death. Although IR therapy may be curative in a number of cancer types, the resistance of cancer cells to radiation remains a major therapeutic problem. In this review, we describe the morphological and molecular aspects of various IR-induced types of cell death. We also discuss cytogenetic variations representative of IR-induced DNA damage and genomic instability. Most importantly, we focus on several pathways and their associated marker proteins responsible for cancer resistance and its therapeutic implications in terms of cancer cell death of various types and characteristics. Finally, we propose radiation-sensitization strategies, such as the modification of fractionation, inflammation, and hypoxia and the combined treatment, that can counteract the resistance of tumors to IR.


Subject(s)
Chromosome Aberrations , Combined Modality Therapy/methods , Gamma Rays/therapeutic use , Neoplasms/therapy , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/therapeutic use , Apoptosis/genetics , Apoptosis/radiation effects , Autophagy/genetics , Autophagy/radiation effects , Cytogenetic Analysis , DNA Damage/radiation effects , Genomic Instability , Humans , Mitosis/radiation effects , Necrosis/genetics , Necrosis/pathology , Neoplasms/genetics , Neoplasms/pathology , X-Ray Therapy
12.
J Biol Chem ; 288(17): 12014-21, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23479728

ABSTRACT

Identification of new anti-apoptotic genes is important for understanding the molecular mechanisms underlying apoptosis and tumorigenesis. The present study identified a novel anti-apoptotic gene named AREL1, which encodes a HECT (homologous to E6-AP carboxyl terminus) family E3 ubiquitin ligase. AREL1 interacted with and ubiquitinated IAP antagonists such as SMAC, HtrA2, and ARTS. However, AREL1 was cytosolic and did not localize to nuclei or mitochondria. The interactions between AREL1 and the IAP antagonists were specific for apoptosis-stimulated cells, in which the IAP antagonists were released into the cytosol from mitochondria. Furthermore, the ubiquitination and degradation of SMAC, HtrA2, and ARTS were significantly enhanced in AREL1-expressing cells following apoptotic stimulation, indicating that AREL1 binds to and ubiquitinates cytosolic but not mitochondria-associated forms of IAP antagonists. Furthermore, the anti-apoptotic role of AREL1-mediated degradation of SMAC, HtrA2, and ARTS was shown by simultaneous knockdown of three IAP antagonists, which caused the inhibition of caspase-3 cleavage, XIAP degradation, and induction of apoptosis. Therefore, the present study suggests that AREL1-mediated ubiquitination and degradation of cytosolic forms of three IAP antagonists plays an important role in the regulation of apoptosis.


Subject(s)
Apoptosis/physiology , Carrier Proteins/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondrial Proteins/metabolism , Proteolysis , Septins/metabolism , Serine Endopeptidases/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/physiology , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins , Carrier Proteins/genetics , Cell Line, Tumor , High-Temperature Requirement A Serine Peptidase 2 , Humans , Inhibitor of Apoptosis Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mitochondrial Proteins/genetics , Molecular Sequence Data , Septins/genetics , Serine Endopeptidases/genetics , Ubiquitin-Protein Ligases/genetics
13.
Biochem Biophys Res Commun ; 445(3): 615-21, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24565837

ABSTRACT

Bacterial lipopolysaccharide (LPS) is an important mediator of inflammation and a potent inducer of endothelial cell damage and apoptosis. In this study, we investigated the protective effects of saikosaponin C (SSc), one of the active ingredients produced by the traditional Chinese herb, Radix Bupleuri, against LPS-induced apoptosis in human umbilical endothelial cells (HUVECs). LPS triggered caspase-3 activation, which was found to be important in LPS-induced HUVEC apoptosis. Inhibition of caspase-3 also inhibited LPS-induced degradation of focal adhesion kinase (FAK), indicating that caspase-3 is important in LPS-mediated FAK degradation as well as in apoptosis in HUVECs. SSc significantly inhibited LPS-induced apoptotic cell death in HUVECs through the selective suppression of caspase-3. SSc was also shown to rescue LPS-induced FAK degradation and other cell adhesion signals. Furthermore, the protective effects of SSc against LPS-induced apoptosis were abolished upon pretreatment with a FAK inhibitor, highlighting the importance of FAK in SSc activity. Taken together, these results show that SSc efficiently inhibited LPS-induced apoptotic cell death via inhibition of caspase-3 activation and caspase-3-mediated-FAK degradation. Therefore, SSc represents a promising therapeutic candidate for the treatment of vascular endothelial cell injury and cellular dysfunction.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Caspase 3/immunology , Focal Adhesion Protein-Tyrosine Kinases/immunology , Human Umbilical Vein Endothelial Cells/drug effects , Lipopolysaccharides/immunology , Oleanolic Acid/analogs & derivatives , Saponins/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Apoptosis/drug effects , Bupleurum/chemistry , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/immunology , Humans , Oleanolic Acid/chemistry , Oleanolic Acid/pharmacology , Saponins/chemistry
14.
Biochem Biophys Res Commun ; 448(3): 267-73, 2014 Jun 06.
Article in English | MEDLINE | ID: mdl-24796672

ABSTRACT

ω-Hydroxyundec-9-enoic acid (ω-HUA), a hydroxyl unsaturated fatty acid derivative, is involved in the antifungal activity of wild rice (Oryza officinalis). Here, we investigated the anti-cancer activity of ω-HUA on a non-small cell lung cancer (NSCLC) cell line. ω-HUA increased apoptosis and induced cleavages of caspase-6, caspase-9, and poly (ADP-ribose) polymerase (PARP). ω-HUA treatment significantly induced endoplasmic reticulum (ER) stress response. Suppression of CHOP expression and inhibiting ER stress by 4-phenylbutyrate (4-PBA) significantly attenuated the ω-HUA treatment-induced activation of caspase-6, caspase-9, and PARP, and subsequent apoptotic cell death, indicating a role for ER stress in ω-HUA-induced apoptosis. In addition, cells subjected to ω-HUA exhibited significantly increased quantity of reactive oxygen species (ROS), and the ROS scavenger N-acetyl-L-cysteine (NAC) inhibited ω-HUA-induced apoptotic cell death and ER stress signals, indicating a role for ROS in ER stress-mediated apoptosis in ω-HUA-treated cells. Taken together, these results suggest that sequential ROS generation and ER stress activation are critical in ω-HUA treatment-induced apoptosis and that ω-HUA represents a promising candidate for NSCLC treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Undecylenic Acids/pharmacology , Acetylcysteine/pharmacology , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Caspase 6/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , Free Radical Scavengers/pharmacology , Gene Knockdown Techniques , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Poly(ADP-ribose) Polymerases/metabolism , Reactive Oxygen Species/metabolism , Transcription Factor CHOP/antagonists & inhibitors , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism
15.
Transl Neurodegener ; 13(1): 4, 2024 01 09.
Article in English | MEDLINE | ID: mdl-38195518

ABSTRACT

Alzheimer's disease (AD) is the most prevalent form of dementia in the elderly and represents a major clinical challenge in the ageing society. Neuropathological hallmarks of AD include neurofibrillary tangles composed of hyperphosphorylated tau, senile plaques derived from the deposition of amyloid-ß (Aß) peptides, brain atrophy induced by neuronal loss, and synaptic dysfunctions. Death-associated protein kinase 1 (DAPK1) is ubiquitously expressed in the central nervous system. Dysregulation of DAPK1 has been shown to contribute to various neurological diseases including AD, ischemic stroke and Parkinson's disease (PD). We have established an upstream effect of DAPK1 on Aß and tau pathologies and neuronal apoptosis through kinase-mediated protein phosphorylation, supporting a causal role of DAPK1 in the pathophysiology of AD. In this review, we summarize current knowledge about how DAPK1 is involved in various AD pathological changes including tau hyperphosphorylation, Aß deposition, neuronal cell death and synaptic degeneration. The underlying molecular mechanisms of DAPK1 dysregulation in AD are discussed. We also review the recent progress regarding the development of novel DAPK1 modulators and their potential applications in AD intervention. These findings substantiate DAPK1 as a novel therapeutic target for the development of multifunctional disease-modifying treatments for AD and other neurological disorders.


Subject(s)
Alzheimer Disease , Aged , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Death-Associated Protein Kinases/genetics , Amyloid beta-Peptides , Central Nervous System , Neurofibrillary Tangles
16.
Arch Pharm Res ; 46(11-12): 882-896, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37804415

ABSTRACT

Breast cancer is one of the major malignancies in women, and most related deaths are due to recurrence, drug resistance, and metastasis. The expression of the mouse double minute 2 (MDM2) oncogene is upregulated in breast cancer; however, its regulatory mechanism has yet to be fully elucidated. Herein, we identified the tumor suppressor death-associated protein kinase 1 (DAPK1) as a novel MDM2 regulator by unbiased peptide library screening. DAPK1 is directly bound to MDM2 and phosphorylates it at Thr419. DAPK1-mediated MDM2 phosphorylation promoted its protein degradation via the ubiquitin-proteasome pathway, resulting in upregulated p53 expression. DAPK1 overexpression, but not its kinase activity-deficient form, decreased colony formation and increased doxorubicin-induced cell death; however, DAPK1 knockdown produced the opposite effects in human breast cancer cells. In a xenograft tumorigenesis assay, DAPK1 overexpression significantly reduced tumor formation, whereas inhibition of DAPK1 kinase activity reduced its antitumorigenic effect. Finally, DAPK1 expression was negatively correlated with MDM2 levels in human breast cancer tissues. Thus, these results suggest that DAPK1-mediated MDM2 phosphorylation and its protein degradation may contribute to its antitumorigenic function in breast cancer.


Subject(s)
Breast Neoplasms , Tumor Suppressor Protein p53 , Animals , Female , Humans , Mice , Breast Neoplasms/drug therapy , Cell Line, Tumor , Death-Associated Protein Kinases/metabolism , Phosphorylation , Protein Stability , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism
17.
Apoptosis ; 17(2): 200-16, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22002103

ABSTRACT

In the present study, we investigated the signaling pathways implicated in the induction of apoptosis by two modified nucleosides, 5-phenylselenyl-methyl-2'-deoxyuridine (PhSe-T) and 5-methylselenyl-methyl-2'-deoxyuridine (MeSe-T), using human cancer cell lines. The induction of apoptosis was associated with proteolytic activation of caspase-3 and -9, PARP cleavage, and decreased levels of IAP family members, including c-IAP-1 and c-IAP-2, but had no effect on XIAP and survivin. PhSe-T and MeSe-T also enhanced the activities of caspase-2 and -8, Bid cleavage, and the conformational activation of Bax. Additionally, nucleoside derivative-induced apoptosis was inhibited by the selective inhibitors of caspase-2, -3, -8, and -9 and also by si-RNAs against caspase-2, -3, -8, and -9; however, inhibition of caspase-2 and -3 was more effective at preventing apoptosis than inhibition of caspase-8 and -9. Moreover, the inhibition of caspase-2 activation by the pharmacological inhibitor z-VDVAD-fmk or by the knockdown of protein expression using siRNA suppressed nucleoside derivative-induced caspase-3 activation, but not vice versa. PhSe-T and MeSe-T also induced a Δψ(m) loss via a CsA-insensitive mechanism, ROS production, and DNA damage, including strand breaks. Moreover, ROS scavengers such as NAC, tiron, and quercetin inhibited nucleoside derivative-induced ROS generation and apoptosis by blocking the sequential activation of caspase-2 and -3, indicating the role of ROS in caspase-2-mediated apoptosis. Taken together, these results indicate that caspase-2 acts upstream of caspase-3 and that caspase-2 functions in response to DNA damage in both PhSe-T- and MeSe-T-induced apoptosis. Our results also suggest that ROS are critical regulators of the sequential activation of caspase-2 and -3 in nucleoside derivative-treated cancer cells.


Subject(s)
Apoptosis/drug effects , Caspase 2/metabolism , Caspase 3/metabolism , Deoxyuridine/analogs & derivatives , Neoplasms/metabolism , Organoselenium Compounds/administration & dosage , Apoptosis/genetics , Caspase 2/genetics , Caspase 3/genetics , Caspase Inhibitors , Cell Line, Tumor , DNA Damage/drug effects , Deoxyuridine/administration & dosage , Enzyme Activation/drug effects , Humans , Oligopeptides/administration & dosage , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction
18.
Biochem Biophys Res Commun ; 417(1): 237-44, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22142847

ABSTRACT

Two novel, modified thymidine nucleosides, 5-phenylselenyl-methyl-2'-deoxyuridine (PhSe-T) and 5-methylselenyl-methyl-2'-deoxyuridine (MeSe-T), trigger reactive oxygen species (ROS) generation and DNA damage and thereby induce caspase-mediated apoptosis in human HL-60 cells; however, the mechanism leading to caspase activation and apoptotic cell death remains unclear. Therefore, we investigated the signaling molecules involved in nucleoside derivative-induced caspase activation and apoptosis in HL-60 cells. PhSe-T/MeSe-T treatment activated two mitogen-activated protein kinases (MAPKs), extracellular-receptor kinase (ERK) and p38, and induced the phosphorylation of two downstream targets of p38, ATF-2 and MAPKAPK2. In addition, the selective p38 inhibitor SB203580 suppressed PhSe-T/MeSe-T-induced apoptosis and activation of caspase-3, -9, -8, and -2, whereas the jun amino-terminal kinase (JNK) inhibitor SP600125 and the ERK inhibitor PD98059 had no effect. SB203580 and an ROS scavenger, tiron, inhibited PhSe-T/MeSe-T-induced histone H2AX phosphorylation, which is a DNA damage marker. Moreover, tiron inhibited PhSe-T/MeSe-T-induced phosphorylation of p38 and enhanced p38 MAP kinase activity, indicating a role for ROS in PhSe-T/MeSe-T-induced p38 activation. Taken together, our results suggest that PhSe-T/MeSe-T-induced apoptosis is mediated by the p38 pathway and that p38 serves as a link between ROS generation and DNA damage/caspase activation in HL-60 cells.


Subject(s)
Apoptosis/drug effects , Deoxyuridine/analogs & derivatives , Organoselenium Compounds/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Caspase 2/biosynthesis , Caspase 3/biosynthesis , Caspase 8/biosynthesis , DNA Damage , Deoxyuridine/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , HL-60 Cells , Humans , Imidazoles/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Pyridines/pharmacology , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
19.
IUBMB Life ; 64(3): 259-65, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22252975

ABSTRACT

The recombinant kringle domain (UK1) of urokinase plasminogen activator was previously reported to exert antiangiogenic activity against Vascular Endothelial Growth Factor (VEGF)-induced angiogenesis in both in vitro and in vivo models. In this study, we explored the molecular signaling mechanisms involved in the antiangiogenic activity of UK1 by examining VEGF signaling proteins. VEGF165 stimulates the phosphorylation of VEGF signaling molecules, and pretreatment with UK1 blocked VEGF-induced signal transduction associated with proliferation, survival, and migration. UK1 also suppressed VEGF165-induced activation of MMP-2. Moreover, UK1 suppressed the phosphorylation and activation of VEGFR2 in VEGF-stimulated human umbilical cord vein endothelial cells (HUVECs) by blocking the dimerization of VEGFR2. Overall, our findings suggest that UK1 inhibits VEGF-induced proliferation, migration, and matrix metalloproteinase activity of HUVECs by suppressing VEGFR2 dimerization and subsequent angiogenic signals.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic , Recombinant Proteins/metabolism , Signal Transduction , Urokinase-Type Plasminogen Activator/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Angiogenesis Inducing Agents , Blotting, Western , Cell Movement , Cell Proliferation , Cells, Cultured , Dimerization , Enzyme-Linked Immunosorbent Assay , Human Umbilical Vein Endothelial Cells/cytology , Humans , Kringles , Matrix Metalloproteinase 2/metabolism , Phosphorylation , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
20.
Apoptosis ; 16(2): 184-97, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21107704

ABSTRACT

In this study, we investigated the signaling pathways implicated in SSa-induced apoptosis of human colon carcinoma (HCC) cell lines. SSa-induced apoptosis of HCC cells was associated with proteolytic activation of caspase-9, caspase-3, and PARP cleavages and decreased levels of IAP family members, such as XIAP and c-IAP-2, but not of survivin. The fluorescence intensity of DiOC6 was significantly reduced after SSa treatment. CsA significantly inhibited SSa-induced loss of mitochondrial transmembrane potential and moderately inhibited SSa-induced cell death. SSa treatment also enhanced the activities of caspase-2 and caspase-8, Bid cleavage, and the conformational activation of Bax. Additionally, SSa-induced apoptosis was inhibited by both the selective caspase-2 inhibitor z-VDVAD-fmk and the selective caspase-8 inhibitor z-IETD-fmk and also by si-RNAs against caspase-2 and caspase-8. The selective caspase-9 inhibitor, z-LEHD-fmk, also inhibited SSa-induced apoptosis, albeit to a lesser extent compared to z-VDVAD-fmk and z-IETD-fmk, indicating that both mitochondria-dependent and mitochondria-independent pathways are associated with SSa-induced apoptosis. Both z-VDVAD-fmk and z-IETD-fmk significantly attenuated the colony-inhibiting effect of SSa. Moreover, inhibition of caspase-2 activation by the pharmacological inhibitor z-VDVAD-fmk, or by knockdown of protein levels using a si-RNA, suppressed SSa-induced caspase-8 activation, Bid cleavage, and the conformational activation of Bax. Although caspase-8 is an initiator caspase like caspase-2, the inhibition of caspase-8 activation by knockdown using a si-RNA did not suppress SSa-induced caspase-2 activation. Altogether, our results suggest that sequential activation of caspase-2 and caspase-8 is a critical step in SSa-induced apoptosis.


Subject(s)
Apoptosis/drug effects , Caspase 2/metabolism , Caspase 8/metabolism , Oleanolic Acid/analogs & derivatives , Saponins/pharmacology , BH3 Interacting Domain Death Agonist Protein/metabolism , Blotting, Western , Caspase 9/metabolism , Cell Line, Tumor , Colonic Neoplasms , Cyclosporine/pharmacology , Flow Cytometry , Humans , Inhibitor of Apoptosis Proteins/metabolism , Mitochondria/metabolism , Oleanolic Acid/metabolism , Oleanolic Acid/pharmacology , Oligopeptides/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , RNA, Small Interfering/pharmacology , Saponins/metabolism , Signal Transduction , Survivin , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL