Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 323
Filter
1.
Cells ; 11(23)2022 Dec 02.
Article in English | MEDLINE | ID: mdl-36497168

ABSTRACT

Muscle fiber composition is associated with physical performance, with endurance athletes having a high proportion of slow-twitch muscle fibers compared to power athletes. Approximately 45% of muscle fiber composition is heritable, however, single nucleotide polymorphisms (SNP) underlying inter-individual differences in muscle fiber types remain largely unknown. Based on three whole genome SNP datasets, we have shown that the rs236448 A allele located near the cyclin-dependent kinase inhibitor 1A (CDKN1A) gene was associated with an increased proportion of slow-twitch muscle fibers in Russian (n = 151; p = 0.039), Finnish (n = 287; p = 0.03), and Japanese (n = 207; p = 0.008) cohorts (meta-analysis: p = 7.9 × 10−5. Furthermore, the frequency of the rs236448 A allele was significantly higher in Russian (p = 0.045) and Japanese (p = 0.038) elite endurance athletes compared to ethnically matched power athletes. On the contrary, the C allele was associated with a greater proportion of fast-twitch muscle fibers and a predisposition to power sports. CDKN1A participates in cell cycle regulation and is suppressed by the miR-208b, which has a prominent role in the activation of the slow myofiber gene program. Bioinformatic analysis revealed that the rs236448 C allele was associated with increased CDKN1A expression in whole blood (p = 8.5 × 10−15) and with greater appendicular lean mass (p = 1.2 × 10−5), whereas the A allele was associated with longer durations of exercise (p = 0.044) reported amongst the UK Biobank cohort. Furthermore, the expression of CDKN1A increased in response to strength (p < 0.0001) or sprint (p = 0.00035) training. Accordingly, we found that CDKN1A expression is significantly (p = 0.002) higher in the m. vastus lateralis of strength athletes compared to endurance athletes and is positively correlated with the percentage of fast-twitch muscle fibers (p = 0.018). In conclusion, our data suggest that the CDKN1A rs236448 SNP may be implicated in the determination of muscle fiber composition and may affect athletic performance.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21 , Genome-Wide Association Study , Muscle Fibers, Skeletal , Muscle Fibers, Slow-Twitch , Humans , Athletes , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/physiology , Muscle Fibers, Skeletal/physiology , Muscle Fibers, Slow-Twitch/physiology
2.
J Orthop Surg Res ; 16(1): 259, 2021 Apr 14.
Article in English | MEDLINE | ID: mdl-33853636

ABSTRACT

BACKGROUND: Osteoarthritis (OA) is a worldwide musculoskeletal disorder. However, disease-modifying therapies for OA are not available. Here, we aimed to characterize the molecular signatures of OA and to identify novel therapeutic targets and strategies to improve the treatment of OA. METHODS: We collected genome-wide transcriptome data performed on 132 OA and 74 normal human cartilage or synovium tissues from 7 independent datasets. Differential gene expression analysis and functional enrichment were performed to identify genes and pathways that were dysregulated in OA. The computational drug repurposing method was used to uncover drugs that could be repurposed to treat OA. RESULTS: We identified several pathways associated with the development of OA, such as extracellular matrix organization, inflammation, bone development, and ossification. By protein-protein interaction (PPI) network analysis, we prioritized several hub genes, such as JUN, CDKN1A, VEGFA, and FOXO3. Moreover, we repurposed several FDA-approved drugs, such as cardiac glycosides, that could be used in the treatment of OA. CONCLUSIONS: We proposed that the hub genes we identified would play a role in cartilage homeostasis and could be important diagnostic and therapeutic targets. Drugs such as cardiac glycosides provided new possibilities for the treatment of OA.


Subject(s)
Computational Biology/methods , Drug Repositioning/methods , Gene Expression/genetics , Osteoarthritis/genetics , Protein Interaction Maps/genetics , Proto-Oncogene Proteins c-jun/physiology , Cardiac Glycosides/therapeutic use , Cyclin-Dependent Kinase Inhibitor p21/physiology , Female , Forkhead Box Protein O3/physiology , Genome-Wide Association Study , Humans , Male , Molecular Targeted Therapy , Osteoarthritis/diagnosis , Osteoarthritis/drug therapy , Transcription Factors , Vascular Endothelial Growth Factor A/physiology
3.
Cell Mol Gastroenterol Hepatol ; 11(5): 1387-1404, 2021.
Article in English | MEDLINE | ID: mdl-33484913

ABSTRACT

BACKGROUND & AIMS: A coordinated stress and regenerative response is important after hepatocyte damage. Here, we investigate the phenotypes that result from genetic abrogation of individual components of the checkpoint kinase 2/transformation-related protein 53 (p53)/cyclin-dependent kinase inhibitor 1A (p21) pathway in a murine model of metabolic liver injury. METHODS: Nitisinone was reduced or withdrawn in Fah-/- mice lacking Chk2, p53, or p21, and survival, tumor development, liver injury, and regeneration were analyzed. Partial hepatectomies were performed and mice were challenged with the Fas antibody Jo2. RESULTS: In a model of metabolic liver injury, loss of p53, but not Chk2, impairs the oxidative stress response and aggravates liver damage, indicative of a direct p53-dependent protective effect on hepatocytes. Cell-cycle control during chronic liver injury critically depends on the presence of both p53 and its downstream effector p21. In p53-deficient hepatocytes, unchecked proliferation occurs despite a strong induction of p21, showing a complex interdependency between p21 and p53. The increased regenerative potential in the absence of p53 cannot fully compensate the surplus injury and is not sufficient to promote survival. Despite the distinct phenotypes associated with the loss of individual components of the DNA damage response, gene expression patterns are dominated by the severity of liver injury, but reflect distinct effects of p53 on proliferation and the anti-oxidative stress response. CONCLUSIONS: Characteristic phenotypes result from the genetic abrogation of individual components of the DNA damage-response cascade in a liver injury model. The extent to which loss of gene function can be compensated, or affects injury and proliferation, is related to the level at which the cascade is interrupted. Accession numbers of repository for expression microarray data: GSE156983, GSE156263, GSE156852, and GSE156252.


Subject(s)
Acute Lung Injury/complications , Carcinogenesis/pathology , Checkpoint Kinase 2/physiology , Cyclin-Dependent Kinase Inhibitor p21/physiology , Liver Neoplasms/pathology , Liver Regeneration , Tumor Suppressor Protein p53/physiology , Animals , Liver Neoplasms/etiology , Liver Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
Biochem Biophys Res Commun ; 532(3): 400-405, 2020 11 12.
Article in English | MEDLINE | ID: mdl-32878707

ABSTRACT

Hypoxia could cause vascular smooth muscle hypertrophy, leading to high pulmonary circulation resistance, pulmonary artery (PA) pressure, even pulmonary arterial hypertension (PAH). Recent studies have demonstrated the ability of mesenchymal stem cell (MSC) to ameliorate PAH but the mechanism was controversial. In this study, we revealed that the growth rate of pulmonary artery smooth muscle cells (PASMCs) treated with hypoxia was significantly increased than normal and showed lower expression of potassium channels. However, cells co-cultured with MSC showed decreased proliferation capability and down-regulated expression of ion channel of PAMSCs. The protein array data showed that the changes of PAMSCs was substantially associated with a high level of tumor necrosis factor alpha (TNFα) secretion from MSC. We further demonstrated that TNFα rescued the cell behavior of PAMSCs through activating the expression of P53 and NF-kB and inducing cell cycle arrest by P21/CDK2/CDK4 downregulation. These findings suggested that MSCs could attenuate abnormal function of PAMSCs by TNFα secretion, which was more or less associated with the beneficial effects of MSC on improving PAH.


Subject(s)
Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/physiopathology , Hypoxia/complications , Hypoxia/physiopathology , Mesenchymal Stem Cells/physiology , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Cycle Checkpoints , Coculture Techniques , Cyclin-Dependent Kinase 2/physiology , Cyclin-Dependent Kinase 4/physiology , Cyclin-Dependent Kinase Inhibitor p21/physiology , Humans , Hypertension, Pulmonary/pathology , Hypoxia/pathology , Mesenchymal Stem Cells/pathology , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/physiology , Proteomics , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Signal Transduction
5.
J Cell Mol Med ; 24(18): 10560-10572, 2020 09.
Article in English | MEDLINE | ID: mdl-32779876

ABSTRACT

Our previous studies found overexpression of Musashi2 (MSI2) conduced to the progression and chemoresistance of pancreatic cancer (PC) by negative regulation of Numb and wild type p53 (wtp53). Now, we further investigated the novel signalling involved with MSI2 in PC. We identified inositol-3-phosphate synthase 1 (ISYNA1) as a novel tumour suppressor regulated by MSI2. High MSI2 and low ISYNA1 expression were prevalently observed in 91 PC tissues. ISYNA1 expression was negatively correlated with MSI2 expression, T stage, vascular permeation and poor prognosis in PC patients. What's more, patients expressed high MSI2 and low ISYNA1 level had a significant worse prognosis. And in wtp53 Capan-2 and SW1990 cells, ISYNA1 was downregulated by p53 silencing. ISYNA1 silencing promoted cell proliferation and cell cycle by inhibiting p21 and enhanced cell migration and invasion by upregulating ZEB-1. However, MSI2 silencing upregulated ISYNA1 and p21 but downregulated ZEB-1, which can be rescued by ISYNA1 silencing. Moreover, reduction of cell migration and invasion resulting from MSI2 silencing was significantly reversed by ISYNA1 silencing. In summary, MSI2 facilitates the development of PC through a novel ISYNA1-p21/ZEB-1 pathway, which provides new gene target therapy for PC.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Cyclin-Dependent Kinase Inhibitor p21/physiology , Intramolecular Lyases/physiology , Neoplasm Proteins/physiology , Pancreatic Neoplasms/pathology , RNA-Binding Proteins/physiology , Signal Transduction/physiology , Zinc Finger E-box-Binding Homeobox 1/physiology , Adult , Aged , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/secondary , Cell Line, Tumor , Cell Movement , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Intramolecular Lyases/antagonists & inhibitors , Intramolecular Lyases/biosynthesis , Intramolecular Lyases/genetics , Kaplan-Meier Estimate , Liver Neoplasms/secondary , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Prognosis , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Tumor Suppressor Protein p53/physiology , Zinc Finger E-box-Binding Homeobox 1/biosynthesis , Zinc Finger E-box-Binding Homeobox 1/genetics
6.
Cell Stress Chaperones ; 25(3): 503-508, 2020 05.
Article in English | MEDLINE | ID: mdl-32253738

ABSTRACT

This study examined the role of cyclin-dependent kinase inhibitor 1a (CDK1A, p21) in response to exogenous stressors during mouse preimplantation embryo development. CDKN1A knockdown (KD) one-cell zygotes were exposed to 39 °C heat stress (HS) for 4 days or irradiated by 1 (1-Gy) or 3 (3-Gy) Gy X-rays, and their developmental competence and gene expression were compared with control embryos. CDKN1A KD and HS did not influence early cleavage or subsequent embryonic development; however, HS delayed cavitation and induced elevated Cdkn1a expression in control embryos. Exposure to 1- or 3-Gy had no effect on development to the morula stage; however, a significant number of morulae failed to develop to the blastocyst stage. Interestingly, under the 1-Gy condition, the blastocyst rate of CDKN1A KD embryos (77.7%) was significantly higher than that of the controls (44.4%). In summary, exposure to cellular stressors resulted in the upregulation of Cdkn1a in embryos exposed to HS or X-ray irradiation, particularly in response to heat stress or low-dose X-ray irradiation, and depleting Cdkn1a mRNA alleviated cell cycle arrest. These findings suggest that CDKN1A plays a vital role in cellular senescence during preimplantation embryo development.


Subject(s)
Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Embryonic Development , Stress, Physiological , Animals , Blastocyst/metabolism , Blastocyst/radiation effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/physiology , DNA Damage , Embryonic Development/radiation effects , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Hot Temperature , Mice , RNA, Messenger/metabolism , Stress, Physiological/genetics , X-Rays
7.
Development ; 147(4)2020 02 17.
Article in English | MEDLINE | ID: mdl-31988189

ABSTRACT

Cellular proliferation is a basic process during organ development, tissue homeostasis and disease progression. Likewise, after injury typically multiple cell lineages respond to various cues and proliferate to initiate repair and/or remodeling of the injured tissue. Unravelling the specific role of proliferation of one cell type and its lineage in the context of the whole organism during tissue regeneration and/or disease progression would provide valuable information on these processes. Here, we report a new genetic system that allows cell proliferation to be inhibited in a tissue-specific manner. We generated Cre- or Dre-inducible p21-GFP (ip21-GFP) transgenic mice that enable experimentally induced permanent cell cycle arrest of specific cell lineages of interest, while genetically marking these cells. This system allows for the inhibition of pathogenic cell proliferation. We found that cardiac fibroblast proliferation inhibition significantly reduced scar formation, and promoted neovascularization and cardiomyocyte survival. Additionally, we found that inhibition of one type of cell proliferation (namely, hepatocytes) induces the lineage conversion of another type cells (i.e. ductal cells) during tissue regeneration. These results validate the use of ip21-GFP mice as a new genetic tool for cell lineage-specific inhibition of cell proliferation in vivo.


Subject(s)
Cell Proliferation , Gene Expression Regulation , Genetic Techniques , Alleles , Animals , Cell Lineage , Cyclin-Dependent Kinase Inhibitor p21/physiology , Female , Fibroblasts/physiology , Green Fluorescent Proteins , Heart/growth & development , Heart/physiology , Hepatocytes/cytology , Hepatocytes/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Infarction/pathology , Myocytes, Cardiac/cytology
8.
Dev Cell ; 52(3): 335-349.e7, 2020 02 10.
Article in English | MEDLINE | ID: mdl-31983631

ABSTRACT

E2F transcription factors control the cytokinesis machinery and thereby ploidy in hepatocytes. If or how these proteins limit proliferation of polyploid cells with extra centrosomes remains unknown. Here, we show that the PIDDosome, a signaling platform essential for caspase-2-activation, limits hepatocyte ploidy and is instructed by the E2F network to control p53 in the developing as well as regenerating liver. Casp2 and Pidd1 act as direct transcriptional targets of E2F1 and its antagonists, E2F7 and E2F8, that together co-regulate PIDDosome expression during juvenile liver growth and regeneration. Of note, whereas hepatocyte aneuploidy correlates with the basal ploidy state, the degree of aneuploidy itself is not limited by PIDDosome-dependent p53 activation. Finally, we provide evidence that the same signaling network is engaged to control ploidy in the human liver after resection. Our study defines the PIDDosome as a primary target to manipulate hepatocyte ploidy and proliferation rates in the regenerating liver.


Subject(s)
Caspase 2/physiology , Death Domain Receptor Signaling Adaptor Proteins/physiology , E2F Transcription Factors/physiology , Hepatocytes/cytology , Liver Regeneration , Polyploidy , Tumor Suppressor Protein p53/physiology , Aneuploidy , Animals , CRADD Signaling Adaptor Protein/physiology , Centrosome , Cyclin-Dependent Kinase Inhibitor p21/physiology , Cytokinesis , Female , Hepatocytes/metabolism , Humans , Male , Mice , Mice, Knockout
9.
Clin Transl Oncol ; 22(4): 503-511, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31218648

ABSTRACT

PURPOSE: The purpose of this study was to evaluate effect of MSCs on CRC cell. METHODS: in this study the MSC was isolated from CRC tissue, its effect on CRC cells was investigated in vivo and vitro, and the underlying mechanism was investigated. RESULTS: In this study we found that MSC-CM could promote colorectal cancer cells escape from senescence both in vitro and in vivo. Further research we demonstrated that MSC-CM acted in colorectal cancer cells senescence through P53/P21 pathway. Next we found that MSC-CM regulate P53 via posttranscription method. CONCLUSION: Collectively, these results reveal that MSCs can help colorectal cancer cells defend against senescence through P53/P21 pathway, which may be a new strategy for colorectal cancer therapy.


Subject(s)
Cellular Senescence/physiology , Colorectal Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p21/physiology , Mesenchymal Stem Cells/physiology , Tumor Suppressor Protein p53/physiology , Animals , Cell Line, Tumor , Cell Proliferation , Female , Humans , Mice , Signal Transduction/physiology
10.
Med Hypotheses ; 133: 109414, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31586810

ABSTRACT

Endometriosis is a common gynecological disorder that affects approximately 6-10% of the female population impairing the quality of life of patients. Several pathophysiologic pathways have been proposed as potential regulators of its severity; however, to date, the processes that trigger the onset and that influence the severity of the disease are not fully understood; hence, leading to disease recurrence in approximately 10-67% of cases. Cyclin-dependent kinase inhibitor 1 (p21/WAF1) is a protein that is a major target of p53 and is related to cell cycle arrest (it regulates transition from the G1 to the S phase) when DNA damage is detected. Its activity has been also linked to the angiogenic potential of tumors as it promotes the expression of various kinases that are responsible for endothelial development and function. Although several articles have underlined the importance of this protein in cancer cell development and tumor growth, there are no relevant data in the field of endometriosis. Indirect evidence suggests, however, that it may be involved in the pathogenesis of endometriosis as it inhibits the activity of various kinases which have been correlated with the course and severity of the disease. The present article investigates the background theory that implies the potential role of cyclin-dependent kinase inhibitor 1 (p21/WAF1) in the pathogenesis of endometriosis. Implications for future research are also provided given that indirect evidence seem to associate downregulation of p21 with decreased growth and invasiveness of human endometrial stromal cells.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/physiology , Endometriosis/etiology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Cycle , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/deficiency , Disease Models, Animal , Disease Progression , Drug Design , Endometriosis/metabolism , Female , Forecasting , Humans , Mice , Mice, Transgenic , Neoplasm Proteins/physiology , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic/physiopathology
11.
Mol Nutr Food Res ; 63(22): e1900629, 2019 11.
Article in English | MEDLINE | ID: mdl-31441212

ABSTRACT

SCOPE: Recent evidence demonstrates that resveratrol (RSV) metabolites, but not free RSV, reach malignant tumors (MT) in breast cancer (BC) patients. Since these metabolites, as detected in MT, do not exert short-term antiproliferative or estrogenic/antiestrogenic activities, long-term tumor-senescent chemoprevention has been hypothesized. Consequently, here, for the first time, whether physiologically relevant RSV metabolites can induce senescence in BC cells is investigated. METHODS AND RESULTS: Human BC MCF-7 (wild-type p53) and MDA-MB-231 (mutant p53), and non-tumorigenic MCF-10A cells are treated with free RSV and physiological-derived metabolites (RSV 3-O-glucuronide, RSV 3-O-sulfate, RSV 4'-O-sulfate, dihydroresveratrol (DH-RSV), and DH-RSV 3-Oglucuronide). Cellular senescence is measured by SA-ß-gal activity and senescence-associated markers (p53, p21Cip1/Waf1 , p16INK4a , and phosphorylation status of retinoblastoma (pRb/tRb)). Although no effect is observed in MDA-MB-231 and normal cells, RSV metabolites induce cellular senescence in MCF-7 cells by reducing their clonogenic capacity and arresting cell cycle at G2 M/S phase, but do not induce apoptosis. Senescence is induced through the p53/p21Cip1/Waf1 and p16INK4a /Rb pathways, depending on the RSV metabolite, and requires ABC transporters, but not estrogen receptors. CONCLUSIONS: These data suggest that RSV metabolites, as found in MT from BC patients, are not de-conjugated to release free RSV, but enter the cells and may exert long-term tumor-senescent chemoprevention.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Breast Neoplasms/drug therapy , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p16/physiology , Cyclin-Dependent Kinase Inhibitor p21/physiology , Resveratrol/metabolism , Retinoblastoma Protein/physiology , Tumor Suppressor Protein p53/physiology , Breast Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Female , Glucuronides/pharmacology , Humans , MCF-7 Cells , Resveratrol/pharmacology , Signal Transduction/physiology , Stilbenes/pharmacology
12.
Cells ; 8(5)2019 05 10.
Article in English | MEDLINE | ID: mdl-31083332

ABSTRACT

Fluoride overexposure is an environmental health hazard and can cause enamel and skeletal fluorosis. Previously we demonstrated that fluoride increased acetylated-p53 and its downstream target p21 in ameloblast-derived LS8 cells. However, p21 function in fluoride toxicity is not well characterized. This study seeks to gain a better understanding of how p53 down-stream mediators, p21 and MDM2, respond to fluoride toxicity. LS8 cells were treated with NaF with/without MG-132 (proteasome inhibitor) or Nutlin-3a (MDM2 antagonist). NaF treatment for 2-6 h increased phospho-p21, which can inhibit apoptosis. However, phospho-p21 and p21 were decreased by NaF at 24 h, even though p21 mRNA was significantly increased at this time point. MG-132 reversed the fluoride-mediated p21 decrease, indicating that fluoride facilitates p21 proteasomal degradation. MG-132 suppressed fluoride-induced caspase-3 cleavage, suggesting that the proteasome plays a pro-apoptotic role in fluoride toxicity. NaF increased phospho-MDM2 in vitro and in mouse ameloblasts in vivo. Nutlin-3a suppressed NaF-mediated MDM2-p21 binding to reverse p21 degradation which increased phospho-p21. This suppressed apoptosis after 24 h NaF treatment. These results suggest that MDM2-mediated p21 proteasomal degradation with subsequent phospho-p21 attenuation contributes to fluoride-induced apoptosis. Inhibition of MDM2-mediated p21 degradation may be a potential therapeutic target to mitigate fluoride toxicity.


Subject(s)
Ameloblasts/drug effects , Ameloblasts/metabolism , Apoptosis/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Proto-Oncogene Proteins c-mdm2 , Sodium Fluoride/toxicity , Ameloblasts/cytology , Animals , Cell Line , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/physiology , Dental Enamel/cytology , Dental Enamel/drug effects , Dental Enamel/metabolism , Imidazoles/pharmacology , Leupeptins/pharmacology , Mice , Mice, Inbred C57BL , Piperazines/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/metabolism
13.
Biol Reprod ; 100(6): 1605-1616, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30927408

ABSTRACT

Dexamethasone (Dex), a corticosteroid hormone, is used during the perinatal period to help fetal lung and other organ development. Conversely, Dex-induced cell proliferation has been associated with accelerated aging. Using primary amnion epithelial cells (AECs) from term, not in labor, fetal membranes, we tested the effects of Dex on cell proliferation, senescence, and inflammation. Primary AECs treated with Dex (100 and 200 nM) for 48 h were tested for cell viability (crystal violet dye exclusion), cell cycle progression and/or type of cell death (flow cytometry), expression patterns of steroid receptors (glucocorticoid receptor, progesterone receptor membrane component 1&2), inflammatory mediators (IL-6 and IL-8), and telomere length (quantitative RT-PCR). Mechanistic mediators of senescence (p38MAPK and p21) were determined by western blot analysis. Dex treatment did not induce AEC proliferation, cell cycle, influence viability, or morphology. However, Dex caused dependent telomere length reduction and p38MAPK-independent but p21-dependent (confirmed by treatment with p21 inhibitor UC2288). Senescence was not associated with an increase in inflammatory mediators, which is often associated with senescence. Co-treatment with RU486 produced DNA damage, cell cycle arrest, and cellular necrosis with an increase in inflammatory mediators. The effect of Dex was devoid of changes to steroid receptors, whereas RU486 increased GR expression. Dex treatment of AECs produced nonreplicative and noninflammatory senescence. Extensive use of Dex during the perinatal period may lead to cellular senescence, contributing to cellular aging associated pathologies during the perinatal and neonatal periods.


Subject(s)
Amnion/cytology , Cellular Senescence/drug effects , Dexamethasone/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/physiology , Amnion/drug effects , Amnion/physiology , Cell Proliferation/drug effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/physiology , Epithelial Cells/cytology , Female , Humans , Pregnancy , Primary Cell Culture , Signal Transduction/drug effects , Telomere/drug effects , Telomere/metabolism , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Respir Res ; 19(1): 200, 2018 Oct 10.
Article in English | MEDLINE | ID: mdl-30305109

ABSTRACT

BACKGROUND: Pulmonary arterial smooth muscle cell (PASMC) proliferation in response to hypoxia plays an important role in the vascular remodelling that occurs in hypoxic pulmonary hypertension. MicroRNAs (miRs) are emerging as important regulators in the progression of pulmonary hypertension. In this study, we investigated whether the expression of miR-17-5p is modulated by hypoxia and is involved in the hypoxia-induced proliferation of PASMCs. METHODS: Human PASMCs were cultured under hypoxic conditions. miR-17-5p expression was determined by real-time RT-PCR. A BrdU incorporation assay and time-lapse recording were utilized to determine cell proliferation and migration. RESULTS: PASMC proliferation was increased by moderate hypoxia (3% oxygen) but was reduced by severe hypoxia (0.1% oxygen) after 48 h. Moderate hypoxia induced miR-17-5p expression. Overexpression of miR-17-5p by transfection with miR-17-5p enhanced cell proliferation and migration in normoxia, whereas knockdown of miR-17-5p with anti-miR-17-5p inhibitors significantly reduced cell proliferation and migration. The expression of miR-17-5p target genes, specifically phosphatase and tensin homologue (PTEN) and cyclin-dependent kinase inhibitor 1 (p21WAF1/Cip1, p21), was reduced under moderate hypoxia in PASMCs. Under normoxia, overexpression of miR-17-5p in PASMCs reduced the expression of PTEN and p21. CONCLUSION: Our data indicate that miR-17-5p might play a significant role in hypoxia-induced pulmonary vascular smooth muscle cell proliferation by regulating multiple gene targets, including PTEN and p21, and that miR-17-5p could be a novel therapeutic target for the management of hypoxia-induced PH.


Subject(s)
Cell Proliferation/physiology , Cyclin-Dependent Kinase Inhibitor p21/physiology , MicroRNAs/physiology , Myocytes, Smooth Muscle/metabolism , PTEN Phosphohydrolase/biosynthesis , Pulmonary Artery/metabolism , Cell Hypoxia/physiology , Cell Movement/physiology , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Humans , MicroRNAs/antagonists & inhibitors , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Pulmonary Artery/cytology , Up-Regulation/physiology
15.
Nucleic Acids Res ; 46(16): 8454-8470, 2018 09 19.
Article in English | MEDLINE | ID: mdl-30085096

ABSTRACT

Long interspersed element-1 (LINE-1, L1) composes ∼17% of the human genome. However, genetic interactions between L1 and human immunodeficiency virus type 1 (HIV-1) remain poorly understood. In this study, we found that HIV-1 suppresses L1 retrotransposition. Notably, HIV-1 Vpr strongly inhibited retrotransposition without inhibiting L1 promoter activity. Since Vpr is known to regulate host cell cycle, we examined the possibility whether Vpr suppresses L1 retrotransposition in a cell cycle dependent manner. We showed that the inhibitory effect of a mutant Vpr (H71R), which is unable to arrest the cell cycle, was significantly relieved compared with that of wild-type Vpr, suggesting that Vpr suppresses L1 mobility in a cell cycle dependent manner. Furthermore, a host cell cycle regulator p21Waf1 strongly suppressed L1 retrotransposition. The N-terminal kinase inhibitory domain (KID) of p21 was required for this inhibitory effect. Another KID-containing host cell cycle regulator p27Kip1 also strongly suppressed L1 retrotransposition. We showed that Vpr and p21 coimmunoprecipitated with L1 ORF2p and they suppressed the L1 reverse transcriptase activity in LEAP assay, suggesting that Vpr and p21 inhibit ORF2p-mediated reverse transcription. Altogether, our results suggest that viral and host cell cycle regulatory machinery limit L1 mobility in cultured cells.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/physiology , HIV-1/physiology , Long Interspersed Nucleotide Elements/genetics , vpr Gene Products, Human Immunodeficiency Virus/physiology , Cell Cycle , Cell Line , Cyclin-Dependent Kinase Inhibitor p27/physiology , Endonucleases/metabolism , Genes, Reporter , Genes, vpr , HIV-1/genetics , Humans , Protein Domains , Proteins/metabolism , RNA Interference , RNA-Directed DNA Polymerase/metabolism , Transcription, Genetic , Virion/metabolism
16.
Int J Biol Sci ; 14(2): 165-177, 2018.
Article in English | MEDLINE | ID: mdl-29483835

ABSTRACT

Werner syndrome (WS) is a rare autosomal recessive progeria disease with genetic instability/cancer predisposition, thus a good model in understanding aging related carcinogenesis. Telomere dysfunction induced cellular senescence is essential in the manifestation of the WS phenotype. Our previous data has shown that p21 (encoded by Cdkn1a gene) could induce cellular senescence and suppress cellular growth of ALT (alternative lengthening of telomere) tumors derived from WS, suggested that p21 might play a key role in maintaining senescence of WS cells. To confirm the role of p21 in suppressing telomere dysfunction induced tumorigenesis, we overexpressed dominant negative protein TRF2ΔBΔM in p21-/- mouse embryonic fibroblasts (MEFs). To further stress the cell, we crossed Wrn-/- mice with p21-/- mice to obtained p21-/-Wrn-/- MEFs, and overexpressed TRF2ΔBΔM in these MEFs to induce telomere dysfunction similar to that in WS cells. Our data showed that, in the context of p21-/- TRF2ΔBΔM, loss of p21 function rescued cellular senescence, and induced p53 mutation, but did not induce tumorigenesis. However, in the set of p21-/-Wrn-/- TRF2ΔBΔM, loss of p21 function induced p53 mutation and tumorigenesis. To further verify the role of p21 in suppressing telomere dysfunction related tumorigenesis, we knocked down p21 in non-tumorigenic immortalized cells derived from WS MEFs (mTerc -/-Wrn-/- ), and found that loss of p21 could induce ALT tumorigenesis, which displayed typical smear pattern of telomere length and arc-shaped telomeric DNA. In another hand, recovering telomerase activity in these MEFs could also induce tumorigenesis without affecting p21 expression level. Together our data suggested that p21 controlled cell cycle regulation played an essential role in suppressing telomere dysfunction-related tumorigenesis. These data also suggested that the genetic context is essential in determining the role of p21 in cancer prevention. Therefore, targeting p21 in the treatment of human degenerative diseases would require a personalized genetic background screen.


Subject(s)
Carcinogenesis/genetics , Cyclin-Dependent Kinase Inhibitor p21/physiology , Werner Syndrome/genetics , Animals , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Knockdown Techniques , Genetic Predisposition to Disease , Genomic Instability , Mice , Tumor Suppressor Protein p53/genetics , Werner Syndrome Helicase/genetics
17.
Oncogene ; 37(17): 2213-2224, 2018 04.
Article in English | MEDLINE | ID: mdl-29379162

ABSTRACT

High grade serous carcinoma (HGSC) is the most common subtype of ovarian cancer and it is now widely accepted that this disease often originates from the fallopian tube epithelium. PAX8 is a fallopian tube lineage marker with an essential role in embryonal female genital tract development. In the adult fallopian tube, PAX8 is expressed in the fallopian tube secretory epithelial cell (FTSEC) and its expression is maintained through the process of FTSEC transformation to HGSC. We now report that PAX8 has a pro-proliferative and anti-apoptotic role in HGSC. The tumor suppressor gene TP53 is mutated in close to 100% of HGSC; in the majority of cases, these are missense mutations that endow the mutant p53 protein with potential gain of function (GOF) oncogenic activities. We show that PAX8 positively regulates the expression of TP53 in HGSC and the pro-proliferative role of PAX8 is mediated by the GOF activity of mutant p53. Surprisingly, mutant p53 transcriptionally activates the expression of p21, which localizes to the cytoplasm of HGSC cells where it plays a non-canonical, pro-proliferative role. Together, our findings illustrate how TP53 mutations in HGSC subvert a normal regulatory pathway into a driver of tumor progression.


Subject(s)
Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p21/physiology , Cystadenocarcinoma, Serous/pathology , Ovarian Neoplasms/pathology , PAX8 Transcription Factor/physiology , Tumor Suppressor Protein p53/physiology , Animals , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cystadenocarcinoma, Serous/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mice , Neoplasm Grading , Ovarian Neoplasms/genetics , Signal Transduction/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
18.
Cancer Lett ; 412: 30-36, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29031586

ABSTRACT

Although the complement C5a/C5aR pathway is suggested to play a critical role in tumor pathogenesis, the underlying mechanism has yet to be fully elucidated. In the present study, we found that in patients with gastric cancer in different clinical stages (from stageⅠto stage Ⅳ), both C5aR and p-PI3K/AKT levels were significantly higher in tumoral tissues than in adjacent non-tumoral tissues. In contrast, p21/p-p21 levels were significantly lower in tumoral tissues than in adjacent non-tumoral tissues. In vitro recombinant C5a administration remarkably promoted p-PI3K/p-AKT expression, but inhibited p21/p-p21 expression. Blockage of C5a/C5aR signaling with a C5aR antagonist reversed the C5a-induced inhibitory effect on p21/p-p21 expression. C5a administration to cells pre-treated with a PI3K inhibitor also prevented this inhibitory effect, suggesting the involvement of the PI3K/AKT signaling pathway in C5a/C5aR-mediated suppression of p21/p-p21 expression. In vivo C5aR antagonist treatment caused significant reduction in tumor growth in mice, accompanied by a remarkable elevation in p21/p-p21 expression and reduction in p-PI3K/AKT activation. These results indicate that the C5a/C5aR pathway promotes gastric cancer pathogenesis by suppressing p21/p-p21 expression via activation of PI3K/AKT signaling.


Subject(s)
Complement C5a/physiology , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/physiology , Signal Transduction/physiology , Stomach Neoplasms/etiology , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/physiology , Down-Regulation , Female , Humans , Male , Mice , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology
19.
BMC Musculoskelet Disord ; 18(1): 435, 2017 Nov 09.
Article in English | MEDLINE | ID: mdl-29121899

ABSTRACT

BACKGROUND: p21(WAF1/CIP1/SDI1), a cyclin dependent kinase inhibitor has been shown to influence cell proliferation, differentiation and apoptosis; but more recently, p21 has been implicated in tissue repair. Studies on p21(-/-) knockout mice have demonstrated results that vary from complete regeneration and healing of tissue to attenuated healing. There have however been no studies that have evaluated the role of p21 inhibition in bone healing and remodeling. METHODS: The current study employs a burr-hole fracture model to investigate bone regeneration subsequent to an injury in a p21-/- mouse model. p21-/- and C57BL/6 mice were subjected to a burr-hole fracture on their proximal tibia, and their bony parameters were measured over 4 weeks via in vivo µCT scanning. RESULTS: p21-/- mice present with enhanced healing from week 1 through week 4. Differences in bone formation and resorption potential between the two mouse models are assessed via quantitative and functional assays. While the µCT analysis indicates that p21-/- mice have enhanced bone healing capabilities, it appears that the differences observed may not be due to the function of osteoblasts or osteoclasts. Furthermore, no differences were observed in the differentiation of progenitor cells (mesenchymal or monocytic) into osteoblasts or osteoclasts respectively. CONCLUSIONS: Therefore, it remains unknown how p21 is regulating enhanced fracture repair and further studies are required to determine which cell type(s) are responsible for this regenerative phenotype.


Subject(s)
Bone Regeneration , Cyclin-Dependent Kinase Inhibitor p21/physiology , Animals , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Osteogenesis , Tibial Fractures , X-Ray Microtomography
20.
J Proteomics ; 151: 2-11, 2017 01 16.
Article in English | MEDLINE | ID: mdl-27394697

ABSTRACT

Epithelial to mesenchymal transition (EMT) is a well-orchestrated process that culminates with loss of epithelial phenotype and gain of a mesenchymal and migratory phenotype. EMT enhances cancer cell invasiveness and drug resistance, favoring metastasis. Dysregulation of transcription factors, signaling pathways, miRNAs and growth factors including EGF, TGF-beta and HGF can trigger EMT. In ovarian cancer, overexpression of the EGFR family is associated with more aggressive clinical behavior. Here, the ovarian adenocarcinoma cell line Caov-3 was induced to EMT with EGF in order to identify specific mechanisms controlled by this process. Caov-3 cells induced to EMT were thoroughly validated and a combination of subcellular proteome enrichment, GEL-LC-MS/MS and SILAC strategy allowed consistent proteome identification and quantitation. Protein network analysis of differentially expressed proteins highlighted regulation of metabolism and cell cycle. Activation of relevant signaling pathways, such as PI3K/Akt/mTOR and Ras/Erk MAPK, in response to EGF-induced EMT was validated. Also, EMT did not affected the proliferation rate of Caov-3 cells, but led to cell cycle arrest in G1 phase regulated by increased levels of p21Waf1/Cip1, independently of p53. Furthermore, a decrease in G1 and G2 checkpoint proteins was observed, supporting the involvement of EGF-induced EMT in cell cycle control. BIOLOGICAL SIGNIFICANCE: Cancer is a complex multistep process characterized by accumulation of several hallmarks including epithelial to mesenchymal transition (EMT), which promotes cellular and microenvironmental changes resulting in invasion and migration to distant sites, favoring metastasis. EMT can be triggered by different extracellular stimuli, including growth factors such as EGF. In ovarian cancer, the most lethal gynecological cancer, overexpression of the EGFR family is associated with more aggressive clinical behavior, increasing mortality rate caused by metastasis. Our proteomic data, together with specific validation of specific cellular mechanisms demonstrated that EGF-induced EMT in Caov-3 cells leads to important alterations in metabolic process (protein synthesis) and cell cycle control, supporting the implication of EGF/EMT in cancer metastasis, cancer stem cell generation and, therefore, poor prognosis for the disease.


Subject(s)
Cell Cycle Checkpoints , Epidermal Growth Factor/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Ovarian Neoplasms/pathology , Proteomics/methods , Cell Line, Tumor , Cell Movement , Chromatography, Liquid , Cyclin-Dependent Kinase Inhibitor p21/physiology , Female , G1 Phase Cell Cycle Checkpoints , Humans , Neoplasm Invasiveness , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...