Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Int J Mol Sci ; 21(8)2020 Apr 16.
Article in English | MEDLINE | ID: mdl-32316320

ABSTRACT

Recent evidence indicates that activation of adenosine monophosphate-activated protein kinase (AMPK), a highly conserved sensor and modulator of cellular energy and redox, regulates cell mitosis. However, the underlying molecular mechanisms for AMPKα subunit regulation of chromosome segregation remain poorly understood. This study aimed to ascertain if AMPKα1 deletion contributes to chromosome missegregation by elevating Polo-like kinase 4 (PLK4) expression. Centrosome proteins and aneuploidy were monitored in cultured mouse embryonic fibroblasts (MEFs) isolated from wild type (WT, C57BL/6J) or AMPKα1 homozygous deficient (AMPKα1-/-) mice by Western blotting and metaphase chromosome spread. Deletion of AMPKα1, the predominant AMPKα isoform in immortalized MEFs, led to centrosome amplification and chromosome missegregation, as well as the consequent aneuploidy (34-66%) and micronucleus. Furthermore, AMPKα1 null cells exhibited a significant induction of PLK4. Knockdown of nuclear factor kappa B2/p52 ameliorated the PLK4 elevation in AMPKα1-deleted MEFs. Finally, PLK4 inhibition by Centrinone reversed centrosome amplification of AMPKα1-deleted MEFs. Taken together, our results suggest that AMPKα1 plays a fundamental role in the maintenance of chromosomal integrity through the control of p52-mediated transcription of PLK4, a trigger of centriole biogenesis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Centrosome/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases/deficiency , AMP-Activated Protein Kinases/genetics , Animals , Cells, Cultured , Chromosome Segregation , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B p52 Subunit/antagonists & inhibitors , NF-kappa B p52 Subunit/genetics , NF-kappa B p52 Subunit/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Up-Regulation
2.
J Biol Chem ; 290(14): 9101-10, 2015 Apr 03.
Article in English | MEDLINE | ID: mdl-25681445

ABSTRACT

Therapeutic benefits offered by tyrosine kinase inhibitors (TKIs), such as gefitinib (Iressa) and erlotinib (Tarceva), are limited due to the development of resistance, which contributes to treatment failure and cancer-related mortality. The aim of this study was to elucidate mechanistic insight into cellular perturbations that accompany acquired gefitinib resistance in lung cancer cells. Several lung adenocarcinoma (LAD) cell lines were screened to characterize epidermal growth factor receptor (EGFR) expression and mutation profile. To circumvent intrinsic variations between cell lines with respect to response to drug treatments, we generated gefitinib-resistant H1650 clone by long-term, chronic culture under gefitinib selection of parental cell line. Isogenic cells were analyzed by microarray, Western blot, flow cytometry, and confocal and transmission electron microscope. We observed that although chronic gefitinib treatment provided effective action against its primary target (aberrant EGFR activity), secondary effects resulted in increased cellular reactive oxygen species (ROS). Gefitinib-mediated ROS correlated with epithelial-mesenchymal transition, as well as striking perturbation of mitochondrial morphology and function. However, gefitinib treatment in the presence of ROS scavenger provided a partial rescue of mitochondrial aberrations. Furthermore, withdrawal of gefitinib from previously resistant clones correlated with normalized expression of epithelial-mesenchymal transition genes. These findings demonstrate that chronic gefitinib treatment promotes ROS and mitochondrial dysfunction in lung cancer cells. Antioxidants may alleviate ROS-mediated resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Mitochondria/drug effects , Quinazolines/pharmacology , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Gefitinib , Humans , Mitochondria/metabolism
3.
Biochim Biophys Acta ; 1853(1): 65-73, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25307521

ABSTRACT

Emerging evidence suggests that activation of adenosine monophosphate-activated protein kinase (AMPK), an energy gauge and redox sensor, controls the cell cycle and protects against DNA damage. However, the molecular mechanisms by which AMPKα isoform regulates DNA damage remain largely unknown. The aim of this study was to determine if AMPKα deletion contributes to cellular hyperproliferation by reducing p21(WAF1/Cip1) (p21) expression thereby leading to accumulated DNA damage. The markers for DNA damage, cell cycle proteins, and apoptosis were monitored in cultured mouse embryonic fibroblasts (MEFs) isolated from wild type (WT, C57BL/6J), AMPKα1, or AMPKα2 homozygous deficient (AMPKα1(-/-), AMPKα2(-/-)) mice by Western blot, flow cytometry, and cellular immunofluorescence staining. Deletion of AMPKα1, the predominant AMPKα isoform, but not AMPKα2 in immortalized MEFs led to spontaneous DNA double-strand breaks (DSB) which corresponded to repair protein p53-binding protein 1 (53BP1) foci formation and subsequent apoptosis. Furthermore, AMPKα1 localizes to chromatin and AMPKα1 deletion down-regulates cyclin-dependent kinase inhibitor, p21, an important protein that plays a role in decreasing the incidence of spontaneous DSB via inhibition of cell proliferation. In addition, AMPKα1 null cells exhibited enhanced cell proliferation. Finally, p21 overexpression partially blocked the cellular hyperproliferation of AMPKα1-deleted MEFs via the inhibition of cyclin-dependent kinase 2 (CDK2). Taken together, our results suggest that AMPKα1 plays a fundamental role in controlling the cell cycle thereby affecting DNA damage and cellular apoptosis.


Subject(s)
AMP-Activated Protein Kinases/physiology , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/physiology , DNA Damage , Animals , Cells, Cultured , Chromosomal Proteins, Non-Histone/physiology , Cyclin-Dependent Kinase Inhibitor p21/analysis , DNA-Binding Proteins/physiology , Fibroblasts/physiology , Mice , Mice, Inbred C57BL , Tumor Suppressor p53-Binding Protein 1
5.
J Biol Chem ; 289(3): 1639-48, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24285539

ABSTRACT

Aberrant receptor tyrosine kinase phosphorylation (pRTK) has been associated with diverse pathological conditions, including human neoplasms. In lung cancer, frequent liver kinase B1 (LKB1) mutations correlate with tumor progression, but potential links with pRTK remain unknown. Heightened and sustained receptor activation was demonstrated by LKB1-deficient A549 (lung) and HeLaS3 (cervical) cancer cell lines. Depletion (siRNA) of endogenous LKB1 expression in H1792 lung cancer cells also correlated with increased pRTK. However, ectopic LKB1 expression in A549 and HeLaS3 cell lines, as well as H1975 activating-EGF receptor mutant lung cancer cell resulted in dephosphorylation of several tumor-enhancing RTKs, including EGF receptor, ErbB2, hepatocyte growth factor receptor (c-Met), EphA2, rearranged during transfection (RET), and insulin-like growth factor I receptor. Receptor abrogation correlated with attenuation of phospho-Akt and increased apoptosis. Global phosphatase inhibition by orthovanadate or depletion of protein tyrosine phosphatases (PTPs) resulted in the recovery of receptor phosphorylation. Specifically, the activity of SHP-2, PTP-1ß, and PTP-PEST was enhanced by LKB1-expressing cells. Our findings provide novel insight on how LKB1 loss of expression or function promotes aberrant RTK signaling and rapid growth of cancer cells.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Neoplasms/enzymology , Protein Serine-Threonine Kinases/biosynthesis , Protein Tyrosine Phosphatases/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Signal Transduction , AMP-Activated Protein Kinase Kinases , HeLa Cells , Humans , Neoplasms/genetics , Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Vanadates/pharmacology
6.
Biochim Biophys Acta ; 1833(6): 1562-71, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23517619

ABSTRACT

Processing of ß-amyloid precursor protein (APP) by ß- and γ-secretases in neurons produces amyloid-ß (Aß), whose excess accumulation leads to Alzheimer's disease (AD). Knowledge on subcellular trafficking pathways of APP and its fragments is important for the understanding of AD pathogenesis. We designed fusion proteins comprising a C-terminal fragment of APP (app) and fluorescent proteins GFP (G) and DsRed (D) to permit the tracking of the fusion proteins and fragments in cells. CAD cells expressing these proteins emitted colocalized green and red fluorescence and produce ectodomains, sGapp and sRapp, and Aß, whose level was reduced by inhibitors of ß- and γ-secretases. The presence of GappR in endosomes was observed via colocalization with Rab5. These observations indicated that the fusion proteins were membrane inserted, transported in vesicles and proteolytically processed by the same mechanism for APP. By attenuating fusion protein synthesis with cycloheximide, individual fluorescent colors from the C-terminus of the fusion proteins appeared in the cytosol which was strongly suppressed by ß-secretase inhibitor, suggesting that the ectodomains exit the cell rapidly (t1/2 about 20min) while the C-terminal fragments were retained longer in cells. In live cells, we observed the fluorescence of the ectodomains located between parental fusion proteins and plasma membrane, suggesting that these ectodomain positions are part of their secretion pathway. Our results indicate that the native ectodomain does not play a decisive role for the key features of APP trafficking and processing and the new fusion proteins may lead to novel insights in intracellular activities of APP.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Amyloidosis/pathology , Green Fluorescent Proteins/metabolism , Luminescent Proteins/metabolism , Neurons/metabolism , Recombinant Fusion Proteins/metabolism , Amyloidosis/metabolism , Animals , Blotting, Western , Cell Survival , Cells, Cultured , Fluorescence , Mice , Neurons/cytology , Peptide Fragments/metabolism , Protein Processing, Post-Translational , Protein Transport , Subcellular Fractions , Red Fluorescent Protein
7.
Am J Pathol ; 182(3): 1021-30, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23306156

ABSTRACT

The aim of this study was to determine the role of AMP-activated protein kinase (AMPK) in lipopolysaccharide (LPS)-induced lung endothelial barrier dysfunction and lung injury in vivo. Both cultured human pulmonary artery endothelial cells (HPAECs) and experimental animals [AMPK subunit α-deficient mice and wild-type (WT) control mice (C57BL/6J)] were used. In cultured HPAECs, LPS increased endothelial permeability in parallel with a decrease in AMPK activity. Consistent with this observation, AMPK activation with the potent AMPK activator 5-aminoimidazole-4-carboxamide-1-d-ribofuranoside (AICAR) attenuated LPS-induced endothelial hyperpermeability in vitro. Intratracheal administration of LPS (1 mg/kg) in WT mice reduced AMPK phosphorylation at Thr172 in lung tissue extracts, increased protein content and cell count in bronchial alveolar lavage fluid, and increased Evans Blue dye infiltration into the lung. These same attributes were similarly enhanced in AMPKα-knockout mice, compared with WT mice. Pretreatment with AICAR reduced these lung injury indicators in LPS-treated WT mice. AMPK activation with AICAR attenuated LPS-induced endothelial hyperpermeability by activating the Rac/Cdc42/PAK pathway, with concomitant inhibition of the Rho pathway, and decreased VE-cadherin phosphorylation at Tyr658. We conclude that AMPK activity supports normal endothelial barrier function and that LPS exposure inhibits AMPK, thereby contributing to endothelial barrier dysfunction and lung injury.


Subject(s)
AMP-Activated Protein Kinases/antagonists & inhibitors , Lung Injury/enzymology , Lung Injury/physiopathology , Lung/enzymology , Lung/physiopathology , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Antigens, CD/metabolism , Blood Vessels/drug effects , Blood Vessels/enzymology , Blood Vessels/pathology , Cadherins/metabolism , Cattle , Cell Membrane Permeability/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/pathology , Enzyme Activation/drug effects , Humans , Inflammation/pathology , Lipopolysaccharides , Lung/drug effects , Lung/pathology , Lung Injury/pathology , Mice , Mice, Inbred C57BL , Myosin Light Chains/metabolism , Phosphoprotein Phosphatases/metabolism , Phosphorylation/drug effects , Phosphothreonine/metabolism , Protein Phosphatase 2C , Protein Serine-Threonine Kinases/metabolism , Ribonucleotides/pharmacology , Signal Transduction/drug effects , cdc42 GTP-Binding Protein/metabolism , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/metabolism
8.
Arterioscler Thromb Vasc Biol ; 33(12): 2800-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24115035

ABSTRACT

OBJECTIVE: Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) are critical events in the progression of several vasculopathologies. Adenosine monophosphate-activated protein kinase (AMPK) has been shown to play a pivotal role in cellular proliferation and migration. However, the roles of AMPK in VSMC migration and its underlying molecular mechanisms remain elusive. APPROACH AND RESULTS: VSMC migration and the neointima formation were studied in cultured mouse VSMCs or in carotid artery ligation of wild-type C57BL/6J mice, AMPKα2, AMPKα1 homozygous-deficient (AMPKα2(-/-), AMPKα1(-/-)) mice. Deletion of AMPKα2, but not AMPKα1, led to increased phosphorylation of both IкB kinase α and its downstream target nuclear factor кB2/p100 at serine 866/870. Consequently, phosphor-p100 at S866/870 bound with E3 ubiquitin ligase ß-transducin repeat-containing protein resulting in the proteolytic processing of the p100 precursor and nuclear factor кB2/p52 induction. Interestingly, acetylation of histone H3 at lysine 56 mediated by histone deacetylase-3 reduction was enhanced significantly in AMPKα2(-/-) VSMCs compared with wild-type or AMPKα1(-/-) VSMCs. Moreover, the augmented association of p52/acetylation of histone H3 at lysine 56 with the promoter of ubiquitin E3 ligase, S-phase kinase-associated protein 2, was shown in AMPKα2(-/-) VSMCs by chromatin immunoprecipitation assay. Furthermore, AMPKα2 deletion caused S-phase kinase-associated protein 2-mediated E-cadherin downregulation. S-Phase kinase-associated protein 2 siRNA abolished the increased migration of AMPKα2(-/-) VSMCs via E-cadherin upregulation. Finally, neointima formation after ligation of carotid artery was increased in AMPKα2(-/-), but not AMPKα1(-/-), mice. CONCLUSIONS: We conclude that deletion of AMPKα2 causes aberrant VSMC migration with accelerated neointima formation in vivo.


Subject(s)
AMP-Activated Protein Kinases/deficiency , Cadherins/metabolism , Carotid Artery Diseases/enzymology , Cell Movement , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , S-Phase Kinase-Associated Proteins/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Acetylation , Animals , Binding Sites , Cadherins/genetics , Carotid Artery Diseases/genetics , Carotid Artery Diseases/pathology , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Down-Regulation , Histone Deacetylases/metabolism , Histones/metabolism , I-kappa B Kinase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , NF-kappa B p52 Subunit/metabolism , Neointima , Phosphorylation , Promoter Regions, Genetic , RNA Interference , S-Phase Kinase-Associated Proteins/genetics , Signal Transduction , Transfection , Up-Regulation , beta-Transducin Repeat-Containing Proteins/metabolism
9.
Acta Biochim Biophys Sin (Shanghai) ; 45(8): 613-21, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23676825

ABSTRACT

Memapsin 2 (BACE1, ß-secretase), a membrane aspartic protease, functions in the cleavage of the type I transmembrane protein, ß-amyloid precursor protein (APP), leading to the production of amyloid ß (Aß) in the brain. Since Aß is closely associated with the pathogenesis of Alzheimer's disease, understanding the biological function, particularly the catalytic activities of memapsin 2, would assist in a better understanding of the disease and the development of its inhibitors. The transmembrane and cytosolic domains of memapsin 2 function in cellular transport and localization, which are important regulatory mechanisms for its activity. The catalytic ectodomain contains a long substrate cleft that is responsible for substrate recognition, specificity, and peptide bond hydrolysis. The substrate cleft accommodates 11 residues of the substrate in separate binding subsites. Besides APP, a number of membrane proteins have been reported to be substrates of memapsin 2. The elucidation for the specificity of these subsites and the amino acid sequences surrounding the memapsin 2 cleavage site in these proteins has led to the establishment of a predictive model that can quantitatively estimate the efficiency of cleavage for any potential substrates. Such tools may be employed for future studies of memapsin 2 about its biological function. Herein, we review the current knowledge on the structure-function relationship of memapsin 2 and its relationship in the biological function.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid Precursor Protein Secretases/physiology , Aspartic Acid Endopeptidases/physiology , Amyloid Precursor Protein Secretases/chemistry , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/chemistry , Aspartic Acid Endopeptidases/metabolism , Catalytic Domain , Humans , Protein Transport , Proteolysis , Structure-Activity Relationship , Substrate Specificity
10.
Int J Mol Sci ; 14(2): 2431-48, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23354477

ABSTRACT

Senescent cells are relatively stable, lacking proliferation capacity yet retaining metabolic activity. In contrast, cancer cells are rather invasive and devastating, with uncontrolled proliferative capacity and resistance to cell death signals. Although tumorigenesis and cellular senescence are seemingly opposite pathological events, they are actually driven by a unified mechanism: DNA damage. Integrity of the DNA damage response (DDR) network can impose a tumorigenesis barrier by navigating abnormal cells to cellular senescence. Compromise of DDR, possibly due to the inactivation of DDR components, may prevent cellular senescence but at the expense of tumor formation. Here we provide an overview of the fundamental role of DDR in tumorigenesis and cellular senescence, under the light of the Yin-Yang concept of Chinese philosophy. Emphasis is placed on discussing DDR outcome in the light of in vivo models. This information is critical as it can help make better decisions for clinical treatments of cancer patients.

11.
Oncotarget ; 7(7): 7970-8, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26701889

ABSTRACT

Neuropilin-1 (NRP-1) has emerged as an important driver of tumor-promoting phenotypes of human malignancies. However, incomplete knowledge exists as to how this single-pass transmembrane receptor mediates pleiotropic tumor-promoting functions. The purpose of this study was to evaluate NRP-1 expression and metastatic properties in 94 endometrial cancer and matching serum specimens and in a lung cancer cell line. We found that NRP-1 expression significantly correlated with increased tumoral expression of vascular endothelial growth factor 2 (VEGFR2) and serum levels of hepatocyte growth factor (HGF) and cell growth-stimulating factor (C-GSF). Tumoral NRP-1 also was positively associated with expression of NEDD9, a pro-metastatic protein. In the highly metastatic lung cancer cell line (H1792), stable LKB1 depletion caused increased migration in vitro and accentuated NRP-1 and NEDD9 expression in vivo. Our findings demonstrate that perturbed expression of these targets correlate with metastatic potential of endometrial and lung tumors, providing clinically-relevant biomarker applications for diagnostic and therapeutic targeting.


Subject(s)
Adenocarcinoma/secondary , Biomarkers, Tumor/metabolism , Cell Movement , Endometrial Neoplasms/pathology , Lung Neoplasms/pathology , Neuropilin-1/metabolism , AMP-Activated Protein Kinase Kinases , Adaptor Proteins, Signal Transducing/metabolism , Adenocarcinoma/metabolism , Aged , Aged, 80 and over , Apoptosis , Blotting, Western , Case-Control Studies , Cell Proliferation , Endometrial Neoplasms/metabolism , Female , Flow Cytometry , Humans , Lung Neoplasms/metabolism , Middle Aged , Neoplasm Grading , Phosphoproteins/metabolism , Prognosis , Protein Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured
12.
J Clin Invest ; 124(10): 4590-602, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25180605

ABSTRACT

After internalization, transmembrane receptors (TMRs) are typically recycled back to the cell surface or targeted for degradation. Efficient TMR trafficking is critical for regulation of several processes, including signal transduction pathways, development, and disease. Here, we determined that trafficking of the angiogenic receptor neuropilin-1 (NRP-1) is abrogated by the liver kinase B1 (LKB1), a serine-threonine kinase of the calcium calmodulin family. We found that aberrant NRP-1 expression in tumor cells from patients with lung adenocarcinoma is associated with decreased levels of LKB1. In cultured lung cells, LKB1 accentuated formation of a complex between NRP-1 and RAB7 in late endosomes. LKB1 specifically bound GTP-bound RAB7, but not a dominant-negative GDP-bound form of RAB7, promoting rapid transfer and lysosome degradation of NRP-1. siRNA-mediated depletion of RAB7 disrupted the transfer of NRP-1 to the lysosome, resulting in recovery of the receptor as well as increased tumor growth and angiogenesis. Together, our findings indicate that LKB1 functions as a RAB7 effector and suppresses angiogenesis by promoting the cellular trafficking of NRP-1 from RAB7 vesicles to the lysosome for degradation. Furthermore, these data suggest that LKB1 and NRP-1 have potential as therapeutic targets for limiting tumorigenesis.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Neovascularization, Pathologic , Neuropilin-1/metabolism , Protein Serine-Threonine Kinases/metabolism , rab GTP-Binding Proteins/metabolism , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Aged , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Female , Humans , Lung/cytology , Lung Neoplasms/metabolism , Lysosomes/metabolism , Male , Mice , Middle Aged , Mutation , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , rab7 GTP-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL