Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
J Am Soc Nephrol ; 32(8): 1987-2004, 2021 08.
Article in English | MEDLINE | ID: mdl-34135081

ABSTRACT

BACKGROUND: Peripheral vascular diseases may induce chronic ischemia and cellular injury distal to the arterial obstruction. Cellular senescence involves proliferation arrest in response to stress, which can damage neighboring cells. Renal artery stenosis (RAS) induces stenotic-kidney dysfunction and injury, but whether these arise from cellular senescenceand their temporal pattern remain unknown. METHODS: Chronic renal ischemia was induced in transgenic INK-ATTAC and wild type C57BL/6 mice by unilateral RAS, and kidney function (in vivo micro-MRI) and tissue damage were assessed. Mouse healthy and stenotic kidneys were analyzed using unbiased single-cell RNA-sequencing. To demonstrate translational relevance, cellular senescence was studied in human stenotic kidneys. RESULTS: Using intraperitoneal AP20187 injections starting 1, 2, or 4 weeks after RAS, selective clearance of cells highly expressing p16Ink4a attenuated cellular senescence and improved stenotic-kidney function; however, starting treatment immediately after RAS induction was unsuccessful. Broader clearance of senescent cells, using the oral senolytic combination dasatinib and quercetin, in C57BL/6 RAS mice was more effective in clearing cells positive for p21 (Cdkn1a) and alleviating renal dysfunction and damage. Unbiased, single-cell RNA sequencing in freshly dissociated cells from healthy and stenotic mouse kidneys identified stenotic-kidney epithelial cells undergoing both mesenchymal transition and senescence. As in mice, injured human stenotic kidneys exhibited cellular senescence, suggesting this process is conserved. CONCLUSIONS: Maladaptive tubular cell senescence, involving upregulated p16 (Cdkn2a), p19 (Cdkn2d), and p21 (Cdkn1a) expression, is associated with renal dysfunction and injury in chronic ischemia. These findings support development of senolytic strategies to delay chronic ischemic renal injury.


Subject(s)
Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Ischemia/physiopathology , Kidney/physiopathology , Renal Insufficiency, Chronic/physiopathology , p21-Activated Kinases/metabolism , Animals , Apoptosis/drug effects , Caspase 8/metabolism , Cellular Senescence/drug effects , Cellular Senescence/genetics , Chronic Disease , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Dasatinib/pharmacology , Disease Models, Animal , Enzyme Activation/drug effects , Epithelial Cells/physiology , Epithelial-Mesenchymal Transition , Gene Expression , Heparin-binding EGF-like Growth Factor/genetics , Humans , Ischemia/etiology , Kidney/blood supply , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Osteopontin/genetics , Protein Kinase Inhibitors/pharmacology , Renal Artery Obstruction/complications , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/pathology , Sequence Analysis, RNA , Single-Cell Analysis , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology , Up-Regulation , p21-Activated Kinases/genetics
2.
Asian Pac J Cancer Prev ; 22(4): 1263-1270, 2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33906321

ABSTRACT

OBJECTIVE: To indicate the effect of Epigallocatechin gallate (EGCG) and Cisplatin (DDP) on proliferation of gastric cancer BGC-823 cells and the relative underlying mechanism. METHODS: Cultured BGC-823 cells were treated by 5 µg/mL DDP, 25 µg/mL EGCG and combined 5 µg/mL DDP with 25 µg/mL EGCG, a blank group was used as control. Cell morphology was observed by 4',6-diamidino-2-phenylindole (DAPI) staining. The ability of cell proliferation was detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT)assay. The cell cloning rate was determined by colony formation assay. The ability of cell migration was detected by cell scratch test. The cell cycle distributions and apoptosis were analyzed by flow cytometry, The expression of p19Arf, p53, p21Cip1 mRNA was determined by RT-qPCR. The protein levels of p19Arf, p53, p21Cip1 were measured by Western blot. RESULTS: Compared with DDP or EGCG treatment alone, EGCG combined with DDP treatment significantly caused nuclear shrinkage, reduced the proliferation rate, the ability of cell clone and migration. EGCG combined with DDP treatment caused cell cycle arrest in G1 phase in BGC-823 cells, increase of apoptosis (21.3%) vs EGCG (7.25%) and DDP (3.86%) single-use group (p <0.01), up-regulated gene and protein expressions of p19Arf, p53, p21Cip1 (p <0.01). CONCLUSION: EGCG can enhance the effect of DDP on inhibiting BGC-823 cell proliferation and inducing apoptosis via activating the p19Arf-p53-p21Cip1 signaling pathway.


Subject(s)
Catechin/analogs & derivatives , Cisplatin/pharmacology , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Stomach Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Catechin/metabolism , Catechin/pharmacology , Cell Culture Techniques , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Signal Transduction/physiology , Stomach Neoplasms/metabolism
3.
Nat Aging ; 1(12): 1107-1116, 2021 12.
Article in English | MEDLINE | ID: mdl-35531351

ABSTRACT

Senescent cells contribute to pathology and dysfunction in animal models1. Their sparse distribution and heterogenous phenotype have presented challenges for detecting them in human tissues. We developed a senescence eigengene approach to identify these rare cells within large, diverse populations of postmortem human brain cells. Eigengenes are useful when no single gene reliably captures a phenotype, like senescence; they also help to reduce noise, which is important in large transcriptomic datasets where subtle signals from low-expressing genes can be lost. Each of our eigengenes detected ~2% senescent cells from a population of ~140,000 single nuclei derived from 76 postmortem human brains with various levels of Alzheimer's disease (AD) pathology. More than 97% of the senescent cells were excitatory neurons and overlapped with tau-containing neurofibrillary tangles (NFTs). Cyclin dependent kinase inhibitor 2D (CDKN2D/p19) was predicted as the most significant contributor to the primary senescence eigengene. RNAscope and immunofluorescence confirmed its elevated expression in AD brain tissue whereby p19-expressing neurons had 1.8-fold larger nuclei and significantly more cells with lipofuscin than p19-negative neurons. These hallmark senescence phenotypes were further elevated in the presence of NFTs. Collectively, CDKN2D/p19-expressing neurons with NFTs represent a unique cellular population in human AD with a senescence phenotype. The eigengenes developed may be useful in future senescence profiling studies as they accurately identified senescent cells in snRNASeq datasets and predicted biomarkers for histological investigation.


Subject(s)
Alzheimer Disease , Neurons , Animals , Humans , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Alzheimer Disease/genetics , Cellular Senescence/genetics , Brain/metabolism
4.
Blood Cells Mol Dis ; 85: 102477, 2020 11.
Article in English | MEDLINE | ID: mdl-32711219

ABSTRACT

Chronic myeloid leukemia (CML) is a kind of myeloproliferative disorder caused by a constitutively active BCR-ABL tyrosine kinase. Tyrosine kinase inhibitors (TKIs), imatinib and its derivatives, have achieved great progress in the treatment of CML. However, many CML patients do not respond to TKIs alone. p19INK4d, a cyclin-dependent kinase inhibitor, plays important roles in proliferation, DNA damage repair, apoptosis and cell differentiation, but its role in CML is unknown. Herein, we found that the expression of p19INK4d in CML patients was significantly lower than that in healthy controls. p19INK4d overexpression inhibits cell proliferation through cell cycle arrest, and cooperates with imatinib to inhibit CML more effectively in vitro and in vivo. Mechanistically, p19INK4d decreased the expression of BCR-ABL and its downstream molecules p-Mek1/2, moreover, the expression of Gli-1, c-myc, MUC1, Shh and TC48 also reduced significantly. Collectively, p19INK4d inhibits proliferation and enhances imatinib efficacy in the treatment of CML. These findings maybe have implications for developing potential targets to increase imatinib sensitivity for CML.


Subject(s)
Antineoplastic Agents/therapeutic use , Cyclin-Dependent Kinase Inhibitor p19/genetics , Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Leukemic , Imatinib Mesylate/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Animals , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Leukemic/drug effects , Humans , Imatinib Mesylate/pharmacology , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice, Nude , Signal Transduction/drug effects
5.
Cell Cycle ; 19(15): 1884-1898, 2020 08.
Article in English | MEDLINE | ID: mdl-32594840

ABSTRACT

In males, aging is accompanied by decline in serum testosterone levels due to impairment of testicular Leydig cells. The polycomb protein BMI1 has recently been identified as an anti-aging factor. In our previous study, BMI1 null mice showed decreased serum testosterone and Leydig cell population, excessive oxidative stress and p16/p19 signaling activation. However, a cause-and-effect relationship between phenotypes and pathways was not investigated. Here, we used the rescue approach to study the role of oxidative stress or p16/p19 in BMI1-mediated steroidogenesis. Our results revealed that treatment with antioxidant NAC, but not down-regulation of p16/p19, largely rescued cell senescence, DNA damage and steroidogenesis in BMI1-deficient mouse MLTC-1 and primary Leydig cells. Collectively, our study demonstrates that BMI1 orchestrates steroidogenesis mainly through maintaining redox homeostasis, and thus, BMI1 may be a novel and potential therapeutic target for treatment of hypogonadism.


Subject(s)
Homeostasis , Leydig Cells/metabolism , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , Steroids/biosynthesis , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Heterocyclic Compounds, 2-Ring/pharmacology , Homeostasis/drug effects , Leydig Cells/drug effects , Male , Mice , Models, Biological , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Testis/metabolism , Testosterone/biosynthesis , Thiazoles/pharmacology
6.
Aging Cell ; 19(2): e13095, 2020 02.
Article in English | MEDLINE | ID: mdl-31880094

ABSTRACT

To determine whether 1,25-dihydroxyvitamin D (1,25(OH)2 D) can exert an anti-osteoporosis role through anti-aging mechanisms, we analyzed the bone phenotype of mice with 1,25(OH)2 D deficiency due to deletion of the enzyme, 25-hydroxyvitamin D 1α-hydroxylase, while on a rescue diet. 1,25(OH)2 D deficiency accelerated age-related bone loss by activating the p16/p19 senescence signaling pathway, inhibiting osteoblastic bone formation, and stimulating osteoclastic bone resorption, osteocyte senescence, and senescence-associated secretory phenotype (SASP). Supplementation of exogenous 1,25(OH)2 D3 corrected the osteoporotic phenotype caused by 1,25(OH)2 D deficiency or natural aging by inhibiting the p16/p19 pathway. The proliferation, osteogenic differentiation, and ectopic bone formation of bone marrow mesenchymal stem cells derived from mice with genetically induced deficiency of the vitamin D receptor (VDR) were significantly reduced by mechanisms including increased oxidative stress, DNA damage, and cellular senescence. We also demonstrated that p16 deletion largely rescued the osteoporotic phenotype caused by 1,25(OH)2 D3 deficiency, whereas 1,25(OH)2 D3 could up-regulate the enzyme Ezh2 via VDR-mediated transcription thereby enriching H3K27me3 and repressing p16/p19 transcription. Finally, we demonstrated that treatment with 1,25(OH)2 D3 improved the osteogenic defects of human BM-MSCs caused by repeated passages by stimulating their proliferation and inhibiting their senescence via the VDR-Ezh2-p16 axis. The results of this study therefore indicate that 1,25(OH)2 D3 plays a role in preventing age-related osteoporosis by up-regulating Ezh2 via VDR-mediated transcription, increasing H3K27me3 and repressing p16 transcription, thus promoting the proliferation and osteogenesis of BM-MSCs and inhibiting their senescence, while also stimulating osteoblastic bone formation, and inhibiting osteocyte senescence, SASP, and osteoclastic bone resorption.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Mesenchymal Stem Cells/drug effects , Osteoporosis/drug therapy , Receptors, Calcitriol/metabolism , Vitamin D/analogs & derivatives , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , Aging/genetics , Animals , Bone and Bones/diagnostic imaging , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/physiopathology , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p19/metabolism , DNA Damage/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Histones/metabolism , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Knockout , Osteocytes/drug effects , Osteocytes/metabolism , Osteogenesis/genetics , Osteoporosis/enzymology , Osteoporosis/metabolism , Osteoporosis/physiopathology , Oxidative Stress/genetics , Receptors, Calcitriol/genetics , Vitamin D/pharmacology , Vitamin D/therapeutic use
7.
Curr Drug Targets ; 21(1): 96-102, 2020.
Article in English | MEDLINE | ID: mdl-31400265

ABSTRACT

Cyclin-dependent kinase inhibitors (CDKIs) are important cell cycle regulators. The CDKI family is composed of the INK4 family and the CIP/KIP family. p19INK4d belongs to the INK4 gene family and is involved in a series of normal physiological activities and the pathogenesis of diseases. Many factors play regulatory roles in the p19INK4d gene expression at the transcriptional and posttranscriptional levels. p19INK4d not only regulates the cell cycle but also plays regulatory roles in apoptosis, DNA damage repair, cell differentiation of hematopoietic cells, and cellular senescence. In this review, the regulatory network of the p19INK4d gene expression and its biological functions are summarized, which provides a basis for further study of p19INK4d as a drug target for disease treatment.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p19/physiology , Animals , Apoptosis , Cell Cycle , Cell Differentiation , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p19/biosynthesis , Cyclin-Dependent Kinase Inhibitor p19/genetics , Cyclin-Dependent Kinase Inhibitor p19/metabolism , DNA Repair , Gene Expression Regulation , Humans , Mice
8.
Free Radic Biol Med ; 138: 23-32, 2019 07.
Article in English | MEDLINE | ID: mdl-31029787

ABSTRACT

AIMS: Oxidative stress is known to induce early replicative senescence. Senescence has been proposed to work as a barrier to immortalization and tumor development. Here, we aimed to evaluate the impact of the loss of peroxisome proliferator activated receptor γ co-activator 1α (PGC-1α), a master regulator of oxidative metabolism and mitochondrial reactive oxygen species (ROS) generation, on replicative senescence and immortalization in mouse embryonic fibroblasts (MEFs). RESULTS: We found that primary MEFs lacking PGC-1α showed higher levels of ROS than wild-type MEFs at all cell passages tested. The elevated production of ROS was associated with higher levels of oxidative DNA damage and the increased formation of DNA double-strand breaks. Evaluation of the induction of DNA repair systems in response to γ-radiation indicated that the loss of PGC-1α also resulted in a small but significant reduction in their activity. DNA damage induced the early activation of senescence markers, including an increase in the number of ß-galactosidase-positive cells, the induction of p53 phosphorylation, and the increase in p16 and p19 protein. These changes were, however, not sufficient to reduce proliferation rates of PGC-1α-deficient MEFs at any cell passage tested. Moreover, PGC-1α-deficient cells escaped replicative senescence. INNOVATION & CONCLUSION: PGC-1α plays an important role in the control of cellular senescence and immortalization.


Subject(s)
Cellular Senescence/radiation effects , DNA Repair , DNA/genetics , Fibroblasts/radiation effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Animals , Biomarkers/metabolism , Cell Proliferation/genetics , Cell Proliferation/radiation effects , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p19/genetics , Cyclin-Dependent Kinase Inhibitor p19/metabolism , DNA/metabolism , DNA Breaks, Double-Stranded/radiation effects , Embryo, Mammalian , Fibroblasts/cytology , Fibroblasts/metabolism , Gamma Rays , Gene Expression Regulation , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondria/radiation effects , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/deficiency , Phosphorylation/radiation effects , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
9.
Int J Biol Sci ; 15(4): 870-881, 2019.
Article in English | MEDLINE | ID: mdl-30906217

ABSTRACT

The polycomb repressor B lymphoma Mo-MLV insertion region 1 (BMI1) is a core composition of polycomb repressive complex 1 (PRC1) and contributes to diverse fundamental cellular processes including cell senescence, apoptosis and proliferation. To investigate the role and mechanism of BMI1 in maintaining normal female reproductive function, we compared the differences in reproductive phenotypes between Bmi1-deficient and wild-type female mice. The Bmi1-deficient female mice were then supplemented with N-acetylcysteine in their drinking water to explore whether antioxidant supplementation could improve reproductive dysfunction caused by BMI1 deficiency. The results revealed that Bmi1 deletion resulted in complete infertility in female mice, estrous cycle disorder, and follicular developmental disorders. The reactive oxygen species levels in the ovarian tissue were increased; the ability of antioxidant enzymes was downregulated; the expression levels of p19 and p53 proteins were significantly upregulated. We also found that oocytes derived from Bmi1-deficient mice could not develop into embryos by in vitro fertilization and in vitro culture of embryos. Furthermore, supplementation with the antioxidant NAC not only improved the reproductive defects caused by Bmi1 deletion, but also largely rescued the ability of Bmi1-deficient oocytes to develop into embryos in vitro. These results indicated that cells lacking Bmi1 resulted in female infertility by activating the p16/p19 signaling pathway, increasing oxidative stress and DNA damage, inhibiting granulosa cell proliferation, and inducing granulosa cell apoptosis. Thus, BMI1 may be a novel potential target for the clinical treatment of female infertility.


Subject(s)
Infertility, Female/metabolism , Infertility, Female/physiopathology , Animals , Apoptosis/physiology , Blotting, Western , Cell Proliferation/physiology , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Female , Flow Cytometry , Granulosa Cells/cytology , Granulosa Cells/metabolism , Immunohistochemistry , Male , Mice , Microscopy, Electron , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Ovary/metabolism , Ovary/physiology , Oxidative Stress/physiology , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Reactive Oxygen Species/metabolism
10.
Article in Chinese | MEDLINE | ID: mdl-30808142

ABSTRACT

Objective: This study aimed to investigate the P19ink4d expression in cochlea of mice model with noise induced hearing loss and the role of P19ink4d in the degeneration of inner ear cells. It also searched for P19ink4d gene alterations in patients with profound sensorineural deafness.Method: CBA/J mice were exposed to broad band noise at 101 dB SPL for 2 hours, auditory brainstem response (ABR) were examined to confirm noise lead to the permanent threshold shift. Immunohistochemical staining, Western blotting, and real-time polymerase chain reaction (PCR) were performed on cochlear tissues, to elucidate changes in P19ink4d expression in mice after noise exposure. For clinical evaluation, 400 children from unrelated families with severe or profound sensorineural hearing loss (SNHL) were recruited, genomic DNA was obtained from the patients and was subjected to DNA microarray to screen mutations in 4 most common genes. The sample that carried none of the common mutant alleles were subjected to PCR and sequenced to detect mutations in P19ink4d gene.Result: The ABR threshold shift of mice in the experimental group significantly increased after noise exposure and was higher than that in the null-noise group. The ABR of 1 day post noise was least among experimental groups and there is no statistical different between ABR of 7 days and 14 days post noise. The missing of outer hair cells occurred after noise exposure, while the inner hair cells hardly miss. It was found that the P19ink4d expression increased significantly in the inner ear cells 3 hours after noise exposure, then recovered in 24 hours. Western blot indicated that the amount of P19ink4d increased transitorily 3-6 h after the noise. However, no mutation existed within the coding exons of P19ink4d in the patients with profound sensorineural deafness.Conclusion: The results support the concept that P19ink4d may play an important role in the pathogenesis and development of noise induced hearing loss.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p19 , Evoked Potentials, Auditory, Brain Stem , Hearing Loss, Noise-Induced , Animals , Auditory Threshold , Brain Stem , Child , Cochlea , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Hearing Loss, Noise-Induced/metabolism , Humans , Mice , Mice, Inbred CBA
11.
Aging Cell ; 17(5): e12827, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30058137

ABSTRACT

Senescent cells accumulate in tissues during aging and are considered to underlie several aging-associated phenotypes and diseases. We recently reported that the elimination of p19ARF -expressing senescent cells from lung tissue restored tissue function and gene expression in middle-aged (12-month-old) mice. The aging of lung tissue increases the risk of pulmonary diseases such as emphysema, and cellular senescence is accelerated in emphysema patients. However, there is currently no direct evidence to show that cellular senescence promotes the pathology of emphysema, and the involvement of senescence in the development of this disease has yet to be clarified. We herein demonstrated that p19ARF facilitated the development of pulmonary emphysema in mice. The elimination of p19ARF -expressing cells prevented lung tissue from elastase-induced lung dysfunction. These effects appeared to depend on reduced pulmonary inflammation, which is enhanced after elastase stimulation. Furthermore, the administration of a senolytic drug that selectively kills senescent cells attenuated emphysema-associated pathologies. These results strongly suggest the potential of senescent cells as therapeutic/preventive targets for pulmonary emphysema.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p19/metabolism , Cytoprotection , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Aniline Compounds/pharmacology , Animals , Bronchoalveolar Lavage Fluid , Cellular Senescence , Diphtheria Toxin/metabolism , Female , Heparin-binding EGF-like Growth Factor/metabolism , Lung/pathology , Mice, Inbred C57BL , Pancreatic Elastase , Sulfonamides/pharmacology , Swine
12.
J Med Food ; 21(7): 678-688, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29851371

ABSTRACT

Schisantherin A (SCA) was evaluated for possible function in restoring the learning and memory impairment induced by D-galactose in mice. ICR mice were treated with D-galactose subcutaneously (220 mg·kg-1), and followed by SCA in different doses (1.25, 2.50 and 5.00 mg·kg-1, administered orally) for 42 days. Effects of SCA on learning and memory were examined by step-through tests and Morris water maze tests. The activity of superoxide dismutase (SOD), the content of malondialdehyde (MDA) in the peripheral blood and hippocampus of mice were assayed by water-soluble tetrazolium-1 (WST-1) and thiobarbituric acid (TBA) methods. The contents of 8 hydroxy deoxy guanosine (8-OHdG) in the hippocampus of mice were detected by immunosorbent assay methods, respectively. Quantitative real-time PCR and Western Blot were respectively used to detect the expression of p19, p53, p21, cyclin D1, CDK4 and RB genes, and the phosphorylation of RB in the hippocampus of mice. We found that SCA significantly improved the learning and memory impairment induced by D-galactose in mice. After SCA treatment, SOD activity was increased and the content of MDA was decreased in both peripheral blood and hippocampus of mice. 8-OHDG content was also decreased in the hippocampus of mice. Furthermore, the expression of p19, p53 and p21 genes was reduced and the expression of cyclin D1 and CDK4 and the phosphorylation of RB protein were elevated in the hippocampus. SCA may improve the learning and memory impairment induced by D-galactose by enhancing the antioxidant capacity, and regulating the expression of p19/p53/p21/cyclinD1/CDK4 genes, and the phosphorylation of RB protein in the hippocampus of mice.


Subject(s)
Cyclooctanes/administration & dosage , Dioxoles/administration & dosage , Drugs, Chinese Herbal/administration & dosage , Learning Disabilities/drug therapy , Lignans/administration & dosage , Memory Disorders/drug therapy , Schisandra/chemistry , Animals , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase Inhibitor p19/genetics , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Galactose/adverse effects , Gene Expression/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Learning Disabilities/genetics , Learning Disabilities/metabolism , Learning Disabilities/psychology , Male , Memory/drug effects , Memory Disorders/genetics , Memory Disorders/metabolism , Memory Disorders/psychology , Mice , Mice, Inbred ICR , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
13.
Cell Transplant ; 27(5): 796-813, 2018 05.
Article in English | MEDLINE | ID: mdl-29808713

ABSTRACT

Increased ß-synuclein (Sncb) expression has been described in the aging visual system. Sncb functions as the physiological antagonist of α-synuclein (Snca), which is involved in the development of neurodegenerative diseases, such as Parkinson's and Alzheimer's diseases. However, the exact function of Sncb remains unknown. The aim of this study was to elucidate the age-dependent role of Sncb in brain microvascular endothelial cells (BMECs). BMECs were isolated from the cortices of 5- to 9-d-old Sprague-Dawley rats and were cultured with different concentrations of recombinant Sncb (rSncb) up to 72 h resembling to some degree age-related as well as pathophysiological conditions. Viability, apoptosis, expression levels of Snca, and the members of phospholipase D2 (Pld2)/ p53/ Mouse double minute 2 homolog (Mdm2)/p19(Arf) pathway, response in RAC-alpha serine/threonine-protein kinase (Akt), and stress-mediating factors such as heme oxygenase (decycling) 1 (Hmox) and Nicotinamide adenine dinucleotide phosphate oxygenase 4 (Nox4) were examined. rSncb-induced effects were confirmed through Sncb small interfering RNA (siRNA) knockdown in BMECs. We demonstrated that the viability decreases, while the rate of apoptosis underly dose-dependent alterations. For example, apoptosis increases in BMECs following the treatment with higher dosed rSncb. Furthermore, we observed a decrease in Snca immunostaining and messenger RNA (mRNA) levels following the exposure to higher rScnb concentrations. Akt was shown to be downregulated and pAkt upregulated by this treatment, which was accompanied by a dose-independent increase in p19(Arf) levels and enhanced intracellular Mdm2 translocation in contrast to a dose-dependent p53 activation. Moreover, Pld2 activity was shown to be induced in rSncb-treated BMECs. The expression of Hmox and Nox4 after Sncb treatment was altered on BEMCs. The obtained results demonstrate dose-dependent effects of Sncb on BMECs in vitro. For example, the p53-mediated and Akt-independent apoptosis together with the stress-mediated response of BMECs related to exposure of higher SNCB concentrations may reflect the increase in Sncb with duration of culture as well as its impact on cell decay. Further studies, expanding on the role of Sncb, may help understand its role in the neurodegenerative diseases.


Subject(s)
Aging/metabolism , Apoptosis , Brain/blood supply , Endothelial Cells/pathology , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , beta-Synuclein/metabolism , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Female , Heme Oxygenase-1/metabolism , Microvessels/pathology , NADPH Oxidase 4/metabolism , Phospholipase D/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , alpha-Synuclein/metabolism
14.
Nan Fang Yi Ke Da Xue Xue Bao ; 38(3): 346-352, 2018 Mar 20.
Article in Chinese | MEDLINE | ID: mdl-29643043

ABSTRACT

OBJECTIVE: To study the effect of Shaoyangzhugu (SYZG) Formula (a formula consisting of 9 traditional Chinese drugs) in delaying the degeneration of articular cartilage and the role p19Arf-p53-p21Cip1 signaling pathway in mediating this effect. METHOD: Thirteen aged cynomolgus monkeys with degenerative knee joints were selected based on X-ray findings, and one of them was randomly selected for pathological observation. The other monkeys were randomized equally into SYZG Formula group (treated with SYZG decoction), ammonia moxime group and saline group. All the monkeys were sacrificed after 8 weeks of treatment with intragastric administration of the drugs or saline. The pathology in the knee joint articular cartilage was observed and the mRNA and protein expressions of p19Arf, p53, and p21Cip1 in the articular cartilage were detected using RT-qPCR and Western blotting. RESULTS: The pathological findings of the articular cartilage in old cynomolgus monkeys were consistent with the characteristics of knee osteoarthritis (KOA). Mankin scores of the cynomolgus monkeys were 7.38∓0.52 in SYZG Formula group, 7.88∓0.83 in ammonia moxime group, and 8.38∓0.74 in saline group, showing a significant difference between SYZG Formula group and saline group (P<0.05). The expressions of p19Arf, p53, and p21Cip1 were the lowest in SYZG Formula group and the highest in saline group with significant differences among the 3 groups (P<0.05). CONCLUSION: SYZG Formula can delay chondrocyte senescence by regulating p19Arf-p53-p21Cip1 signaling pathway to delay articular cartilage degeneration in aged cynomolgus monkeys.


Subject(s)
Cartilage, Articular/drug effects , Drugs, Chinese Herbal/pharmacology , Osteoarthritis, Knee/drug therapy , Signal Transduction , Animals , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Macaca fascicularis , Random Allocation , Tumor Suppressor Protein p53/metabolism
15.
Proc Natl Acad Sci U S A ; 115(13): 3344-3349, 2018 03 27.
Article in English | MEDLINE | ID: mdl-29531090

ABSTRACT

Cell cycle progression is tightly regulated by cyclin-dependent kinases (CDKs). The ankyrin-repeat protein p19INK4d functions as a key regulator of G1/S transition; however, its molecular mode of action is unknown. Here, we combine cell and structural biology methods to unravel the mechanism by which p19INK4d controls cell cycle progression. We delineate how the stepwise phosphorylation of p19INK4d Ser66 and Ser76 by cell cycle-independent (p38) and -dependent protein kinases (CDK1), respectively, leads to local unfolding of the three N-terminal ankyrin repeats of p19INK4d This dissociates the CDK6-p19INK4d inhibitory complex and, thereby, activates CDK6. CDK6 triggers entry into S-phase, whereas p19INK4d is ubiquitinated and degraded. Our findings reveal how signaling-dependent p19INK4d unfolding contributes to the irreversibility of G1/S transition.


Subject(s)
Cell Cycle/physiology , Cyclin-Dependent Kinase Inhibitor p19/chemistry , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Protein Unfolding , Cell Division , HEK293 Cells , HeLa Cells , Humans , Models, Molecular , Phosphorylation , Protein Conformation , Proteolysis , Signal Transduction
16.
Cell Prolif ; 51(3): e12447, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29484736

ABSTRACT

OBJECTIVES: Histone deacetylases (HDACs) are commonly dysregulated in cancer and represent promising therapeutic targets. However, global HDAC inhibitors have shown limited efficacy in the treatment of solid tumours, including hepatocellular carcinoma (HCC). In this study, we investigated the therapeutic effect of selectively inhibiting HDAC1 and 2 in HCC. METHODS: HDAC1 inhibitor Tacedinaline (CI994), HDAC2 inhibitor Santacruzamate A (CAY10683), HDAC1/2 common inhibitor Romidepsin (FK228) and global HDAC inhibitor Vorinostat (SAHA) were used to treat HCC cells. Cell cycle, apoptosis and the protein levels of CDKs and CDKNs were performed to evaluate HCC cell growth. Inhibition of HDAC1/2 by RNAi was further investigated. RESULTS: Combined inhibition of HDAC1/2 led to HCC cell morphology changes, growth inhibition, cell cycle blockage and apoptosis in vitro and suppressed the growth of subcutaneous HCC xenograft tumours in vivo. p21Waf1/Cip1 and p19INK4d , which play roles in cell cycle blockage and apoptosis induction, were upregulated. Inhibition of HDAC1/2 by siRNA further demonstrated that HDAC1 and 2 cooperate in blocking the cell cycle and inducing apoptosis via p19INK4d and p21Waf1/Cip1 upregulation. Finally, H3K18, H3K56 and H4K12 in the p19INK4d and p21Waf1/Cip1 promoter regions were found to be targets of HDAC1/2. CONCLUSIONS: Pharmacological or transcriptional inhibition of HDAC1/2 increases p19INK4d and p21Waf1/Cip1 expression, decreases CDK expression and arrests HCC growth. These results indicated a potential pharmacological mechanism of selective HDAC1/2 inhibitors in HCC therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Depsipeptides/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Liver Neoplasms/drug therapy , Animals , Apoptosis , Carcinoma, Hepatocellular/enzymology , Cell Cycle Checkpoints/drug effects , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Humans , Liver Neoplasms/enzymology , Mice, Nude , Transcription, Genetic , Up-Regulation , Vorinostat , Xenograft Model Antitumor Assays
17.
Curr Biol ; 27(21): 3302-3314.e6, 2017 Nov 06.
Article in English | MEDLINE | ID: mdl-29112861

ABSTRACT

Mesothelioma is a fatal tumor of the pleura and is strongly associated with asbestos exposure. The molecular mechanisms underlying the long latency period of mesothelioma and driving carcinogenesis are unknown. Moreover, late diagnosis means that mesothelioma research is commonly focused on end-stage disease. Although disruption of the CDKN2A (INK4A/ARF) locus has been reported in end-stage disease, information is lacking on the status of this key tumor suppressor gene in pleural lesions preceding mesothelioma. Manufactured carbon nanotubes (CNTs) are similar to asbestos in terms of their fibrous shape and biopersistent properties and thus may pose an asbestos-like inhalation hazard. Here we show that instillation of either long CNTs or long asbestos fibers into the pleural cavity of mice induces mesothelioma that exhibits common key pro-oncogenic molecular events throughout the latency period of disease progression. Sustained activation of pro-oncogenic signaling pathways, increased proliferation, and oxidative DNA damage form a common molecular signature of long-CNT- and long-asbestos-fiber-induced pathology. We show that hypermethylation of p16/Ink4a and p19/Arf in CNT- and asbestos-induced inflammatory lesions precedes mesothelioma; this results in silencing of Cdkn2a (Ink4a/Arf) and loss of p16 and p19 protein, consistent with epigenetic alterations playing a gatekeeper role in cancer. In end-stage mesothelioma, silencing of p16/Ink4a is sustained and deletion of p19/Arf is detected, recapitulating human disease. This study addresses the long-standing question of which early molecular changes drive carcinogenesis during the long latency period of mesothelioma development and shows that CNT and asbestos pose a similar health hazard.


Subject(s)
Asbestos/toxicity , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Mesothelioma/chemically induced , Mesothelioma/pathology , Nanotubes, Carbon/toxicity , Aged , Animals , Carcinogenesis/chemically induced , Carcinogenesis/genetics , Cell Proliferation/drug effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p19/genetics , Female , Humans , Lung Neoplasms/genetics , Male , Mesothelioma/genetics , Mesothelioma, Malignant , Methylation/drug effects , Mice , Mice, Inbred C57BL , Middle Aged
18.
Sci Rep ; 7(1): 16577, 2017 11 29.
Article in English | MEDLINE | ID: mdl-29185453

ABSTRACT

YAP is a downstream nuclear transcription factor of Hippo pathway which plays an essential role in development, cell growth, organ size and homeostasis. It was previously identified that elevation of YAP in genomics of genetic engineered mouse (GEM) model of prostate cancer is associated with Pten/Trp53 inactivation and ARF elevation hypothesizing the essential crosstalk of AKT/mTOR/YAP with ARF in prostate cancer. However, the detailed function and trafficking of YAP in cancer cells remains unclear. Using GEM microarray model, we found ARF dysregulates Hippo and Wnt pathways. In particular, ARF knockdown reduced non-nuclear localization of YAP which led to an increase in F-actin. Mechanistically, ARF knockdown suppressed protein turnover of ß-catenin/YAP, and therefore enhanced the activity of AKT and phosphorylation of YAP. Moreover, we found tea-derived carbon dots can interact with ARF in nucleus that may further lead to the non-nuclear localization of YAP. Thus, we reported a novel crosstalk of ARF/ß-catenin dysregulated YAP in Hippo pathway and a new approach to stimulate ARF-mediated signaling to inhibit nuclear YAP using nanomaterials implicating an innovative avenue for treatment of cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carbon/chemistry , Nanotechnology/methods , Phosphoproteins/metabolism , Tea/chemistry , Animals , Cell Cycle Proteins , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p19/genetics , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/physiology , Hippo Signaling Pathway , Male , Mice , Microscopy, Confocal , Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology , YAP-Signaling Proteins
19.
Ann Clin Lab Sci ; 47(5): 523-528, 2017 Sep.
Article in English | MEDLINE | ID: mdl-29066476

ABSTRACT

Medullary thyroid carcinoma (MTC), an aggressive form of thyroid cancer, occurs sporadically in approximately 75% of MTCs. RET and RAS mutations play a role in about 40% and 15%, respectively, of sporadic MTCs and are predominant drivers in MTC pathways. These mutations are some of the most comprehensively described and screened for in MTC patients; however, in recent studies, other mutations in the CDKN2C gene (p18) have been implicated in the tumorigenesis of MTC. Comparative genomic hybridization analysis revealed that approximately 40% of sporadic MTC samples have loss of CDKN2C at chromosome 1p32 in addition to frequent losses of CDKN2D (p19) at chromosome 19p13. However, no feasible routine method had been established to detect loss of heterozygosity (LOH) of CDKN2C and CD-KN2D The aim of this study is to assess the feasibility of using Fluorescence in situ Hybridization (FISH) to screen MTC patients for CDKN2C and CDKN2D deletions. We subjected 5 formalin-fixed, paraffin-embedded (FFPE) MTC samples with defined RET/RAS mutations to dual-color FISH assays to detect loss of CDKN2C and/or CDKN2D We prepared spectrum orange probes using the bacterial artificial chromosomes RP11-779F9 for CDKN2C (p18) and RP11-177J4 for CDKN2D (p19) and prepared spectrum green control probes to the 1q25.2 and 19q11 regions (RP11-1146A3 and RP11-942P7, respectively). Nine FFPE normal thyroid tissue samples were used to establish the cutoff values for the FISH signal patterns. Of the five FFPE MTC samples, four and one yielded a positive significant result for CDKNN2C loss and CDKN2D loss, respectively. The results of a Clinical Laboratory Improvement Amendments validation with a CDKN2C/CKS1B probe set for CDKN2C (p18) loss of heterozygosity were 100% concordant with the FISH results obtained in this study. Thus, FISH is a fast and reliable diagnostic or prognostic indicator of gene loss in MTC.


Subject(s)
Carcinoma, Neuroendocrine/genetics , Cyclin-Dependent Kinase Inhibitor p18/genetics , Cytogenetic Analysis/methods , Neoplasm Proteins/genetics , Thyroid Gland/metabolism , Thyroid Neoplasms/genetics , Cancer Care Facilities , Carcinoma, Neuroendocrine/diagnosis , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/pathology , Chromosomes, Artificial, Bacterial/metabolism , Cyclin-Dependent Kinase Inhibitor p18/metabolism , Cyclin-Dependent Kinase Inhibitor p19/genetics , Cyclin-Dependent Kinase Inhibitor p19/metabolism , DNA, Complementary/metabolism , DNA, Recombinant/metabolism , Feasibility Studies , Gene Deletion , Humans , In Situ Hybridization, Fluorescence , Loss of Heterozygosity , Neoplasm Proteins/metabolism , Prognosis , Recombinant Proteins/metabolism , Reproducibility of Results , Texas , Thyroid Gland/pathology , Thyroid Neoplasms/diagnosis , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Time Factors
20.
J Am Heart Assoc ; 6(7)2017 Jul 19.
Article in English | MEDLINE | ID: mdl-28724653

ABSTRACT

BACKGROUND: Cell senescence is involved in the process of organ damage and repair; however, the underlying molecular mechanism needs to be further explored. METHODS AND RESULTS: Senescence-related genes (ie, p21, p53, and ataxia telangiectasia mutated [ATM]) were shown to be elevated after myocardial infarction (MI) in both mouse and human hearts. Ten- to 12-week-old male wild-type littermates (ATM+/+) and ATM heterozygous mice (ATM+/-) were subjected to MI. Cardiac echography showed that ATM haplodeficiency did not affect the survival rate but aggravated heart failure at day 28 post MI. Histologic analysis showed increased fibrosis in the noninfarct area of ATM+/- mice compared with that in ATM+/+ mice. Senescence-associated ß-galactosidase staining showed that the number of senescent fibroblasts was decreased when ATM was haplodeficient both in vivo and in vitro. Costaining of α-smooth muscle actin with p53 or p19 showed fewer senescent myofibroblasts in ATM+/- mouse hearts. Moreover, angiogenesis was also examined using the endothelial markers CD31 both at early (day 7) and late stages (day 28) after MI, and ATM haplodeficiency reduced angiogenesis after MI. Finally, cardiac fibroblasts were isolated from infarcted mouse heart and the medium were tested for its capacity of endothelial tubing formation, revealing that ATM haplodeficiency led to lower vascular endothelial growth factor production from cardiac fibroblast and reduced capacity of endothelial tube formation in vitro. CONCLUSIONS: The present study shows that ATM haplodeficiency decreases fibroblast senescence and vascular endothelial growth factor production and impaired angiogenesis in response to MI, leading to accelerated heart failure.


Subject(s)
Haploinsufficiency , Heart Failure/genetics , Myocardial Infarction/genetics , Actins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins/genetics , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Disease Models, Animal , Disease Progression , Fibrosis , Genetic Predisposition to Disease , Heart Failure/enzymology , Heart Failure/pathology , Heart Failure/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/enzymology , Myocardium/pathology , Myofibroblasts/enzymology , Myofibroblasts/pathology , Neovascularization, Physiologic , Phenotype , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Signal Transduction , Time Factors , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factor A/metabolism , Ventricular Function, Left , Ventricular Remodeling
SELECTION OF CITATIONS
SEARCH DETAIL