Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 150
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Antimicrob Agents Chemother ; 68(3): e0107223, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38319085

ABSTRACT

Current antiretroviral therapy for HIV-1 infection does not represent a cure for infection as viral rebound inevitably occurs following discontinuation of treatment. The "block and lock" therapeutic strategy is intended to enforce proviral latency and durably suppress viremic reemergence in the absence of other intervention. The transcription-associated cyclin-dependent protein kinases (tCDKs) are required for expression from the 5´ HIV-1 long-terminal repeat, but the therapeutic potential of inhibiting these kinases for enforcing HIV-1 latency has not been characterized. Here, we expanded previous observations to directly compare the effect of highly selective small molecule inhibitors of CDK7 (YKL-5-124), CDK9 (LDC000067), and CDK8/19 (Senexin A), and found each of these prevented HIV-1 provirus expression at concentrations that did not cause cell toxicity. Inhibition of CDK7 caused cell cycle arrest, whereas CDK9 and CDK8/19 inhibitors did not, and could be continuously administered to establish proviral latency. Upon discontinuation of drug administration, HIV immediately rebounded in cells that had been treated with the CDK9 inhibitor, while proviral latency persisted for several days in cells that had been treated with CDK8/19 inhibitors. These results identify the mediator kinases CDK8/CDK19 as potential "block and lock" targets for therapeutic suppression of HIV-1 provirus expression.


Subject(s)
HIV-1 , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinases/pharmacology , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Cyclins/metabolism , Cyclins/pharmacology
2.
Environ Toxicol ; 39(1): 75-84, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37638803

ABSTRACT

In this study, we have investigated the chemopreventive role of 6-shogaol (6-SGL) on benzopyrene (BaP) exposed lung carcinogenesis by modulating PRDX1-associated oxidative stress, inflammation, and proliferation in Swiss albino mouse models. Mice were exposed to BaP (50 mg/kg b.wt) orally twice a week for four consecutive weeks and maintained for 16 weeks, respectively. 6-SGL (30 mg/kg b.wt) were orally administered to mouse 1 h before BaP exposure for 16 weeks. After the experiment's termination, 6-SGL (30 mg/kg b.wt) prevented the loss in body weight, increased lung weight, and the total number of tumors in the mice. Moreover, we observed that 6-SGL treatment reverted the activity of BaP-induced lipid peroxidation and antioxidants in mice. Also, 6-SGL impeded the phosphorylation of MAPK family proteins such as Erk1, p38, and Jnk1 in BaP-exposed mice. PRDX1 is an essential antioxidant protein that scavenges toxic radicals and enhances several antioxidant proteins. Overexpression of PRDX1 substantially inhibits MAPKs, proliferation, and inflammation signaling axis. Hence, PRDX1 is thought to be a novel targeting protein for preventing BaP-induced lung cancer. In this study, we have obtained the 6-SGL treatment in a mouse model that reverted BaP-induced depletion of PRDX1 expression. Moreover, pretreatment of 6-SGL (30 mg/kg b.wt) significantly inhibited enhanced proinflammatory cytokines (TNF-α, IL-6, IL-ß1, IL-10) and proliferative markers (Cyclin-D1, Cyclin-D2, and PCNA) in BaP-exposed mice. The histopathological studies also confirmed that 6-SGL effectively protected the cells with less damage. Thus, the study demonstrated that 6-SGL could be a potential phytochemical and act as a chemopreventive agent in BaP-induced lung cancer by enhancing PRDX1 expression.


Subject(s)
Antioxidants , Lung Neoplasms , Mice , Animals , Antioxidants/metabolism , Benzo(a)pyrene/toxicity , Oxidative Stress , Lung , Carcinogenesis , Inflammation/metabolism , Lung Neoplasms/chemically induced , Lung Neoplasms/prevention & control , Lung Neoplasms/metabolism , Disease Models, Animal , Cell Proliferation , Cyclins/metabolism , Cyclins/pharmacology
3.
Drug Dev Res ; 84(5): 907-921, 2023 08.
Article in English | MEDLINE | ID: mdl-37070571

ABSTRACT

BACKGROUND: Nonsmall cell lung cancer (NSCLC) is the main type of lung cancer, accounting for approximately 85%. Berberine (BBR), a commonly used traditional Chinese medicine, has been reported to exhibit a potential antitumor effect in various cancers. In this research, we explored the function of BBR and its underlying mechanisms in the development of NSCLC. METHODS: Cell Counting Kit-8 (CCK-8), 5-ethynyl-20-deoxyuridine (EdU), colony formation assays, flow cytometry, and transwell invasion assay were employed to determine cell growth, the apoptotic rate, cell invasion of NSCLC cells, respectively. Western blot was applied for detecting the protein expression of c-Myc, matrix metalloprotease 9 (MMP9), kinesin family member 20A (KIF20A), cyclin E2 (CCNE2), and phosphatidylinositol-3-kinase/protein kinase B (PI3K/AKT) pathway-related proteins. Glycolysis was evaluated by detecting glucose consumption, lactate production, and adenosine triphosphate/adenosine diphosphate (ATP/ADP) ratio with the matched kits. Real-time quantitative polymerase chain reaction (RT-qPCR) was conducted to analyze the level of KIF20A and CCNE2. Tumor model was established to evaluate the function of BBR on tumor growth in NSCLC in vivo. In addition, immunohistochemistry assay was employed to detect the level of KIF20A, CCNE2, c-Myc, and MMP9 in mice tissues. RESULTS: BBR exhibited suppressive effects on the progression of NSCLC, as evidenced by inhibiting cell growth, invasion, glycolysis, and facilitating cell apoptosis in H1299 and A549 cells. KIF20A and CCNE2 were upregulated in NSCLC tissues and cells. Moreover, BBR treatment significantly decreased the expression of KIF20A and CCNE2. KIF20A or CCNE2 downregulation could repress cell proliferation, invasion, glycolysis, and induce cell apoptosis in both H1299 and A549 cells. The inhibition effects of BBR treatment on cell proliferation, invasion, glycolysis, and promotion effect on cell apoptosis were rescued by KIF20A or CCNE2 overexpression in NSCLC cells. The inactivation of PI3K/AKT pathway caused by BBR treatment in H1299 and A549 cells was restored by KIF20A or CCNE2 upregulation. In vivo experiments also demonstrated that BBR treatment could repress tumor growth by regulating KIF20A and CCNE2 and inactivating the PI3K/AKT pathway. CONCLUSION: BBR treatment showed the suppressive impact on the progression of NSCLC by targeting KIF20A and CCNE2, thereby inhibiting the activation of the PI3K/AKT pathway.


Subject(s)
Berberine , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Mice , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Berberine/pharmacology , Matrix Metalloproteinase 9 , Signal Transduction , Cell Proliferation , Apoptosis , Cyclins/metabolism , Cyclins/pharmacology , Cell Line, Tumor , Cell Movement
4.
J Med Virol ; 94(6): 2384-2387, 2022 06.
Article in English | MEDLINE | ID: mdl-34964486

ABSTRACT

More than 20 members of the human cyclin-dependent kinases (CDKs) family share the feature of being activated by cyclins. CDKs have been involved in diverse biological processes, such as cell cycle, transcription, DNA damage response, and apoptosis. If CDKs are not properly regulated, they can cause diseases like cancer. CDKs are Ser/Thr kinases that work with cyclins to control cell cycle progression. Various CDK-cyclin complexes phosphorylate particular target proteins and drive different cell cycle stages. Accumulating evidence demonstrated that CDKs play an essential role in the cell cycle; however, their roles in antiviral innate immunity are just emerging. This minireview summarizes how CDKs play their roles in antiviral innate immunity. Our goal is to draw attention to the involvement of CDKs in antiviral innate immunity, whether as separate entities or as components of CDK/cyclin complexes that have gotten less attention in the past.


Subject(s)
Antiviral Agents , Cyclin-Dependent Kinases , Antiviral Agents/pharmacology , Cell Cycle , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Cyclins/pharmacology , Humans , Immunity, Innate
5.
J Appl Biomed ; 19(2): 113-124, 2021.
Article in English | MEDLINE | ID: mdl-34754259

ABSTRACT

Oplopanax elatus (Nakai) Nakai has a long history of use as an ethnomedicine by the people living in eastern Asia. However, its bioactive constituents and cancer chemopreventive mechanisms are largely unknown. The aim of this study was to prepare O. elatus extracts, fractions, and single compounds and to investigate the herb's antiproliferative effects on colon cancer cells and the involved mechanisms of action. Two polyyne compounds were isolated from O. elatus, falcarindiol and oplopandiol. Based on our HPLC analysis, falcarindiol and oplopandiol are major constituents in the dichloromethane (CH2Cl2) fraction. For the HCT-116 cell line, the dichloromethane fraction showed significant effects. Furthermore, the IC50 for falcarindiol and oplopandiol was 1.7 µM and 15.5 µM, respectively. In the mechanistic study, after treatment with 5 µg/ml for 48 h, dichloromethane fraction induced cancer cell apoptosis by 36.5% (p < 0.01% vs. control of 3.9%). Under the same treatment condition, dichloromethane fraction caused cell cycle arrest at the G2/M phase by 32.6% (p < 0.01% vs. control of 23.4%), supported by upregulation of key cell cycle regulator cyclin A to 21.6% (p < 0.01% vs. control of 8.6%). Similar trends were observed by using cell line HT-29. Data from this study filled the gap between phytochemical components and the cancer chemoprevention of O. elatus. The dichloromethane fraction is a bioactive fraction, and falcarindiol is identified as an active constituent. The mechanisms involved in cancer chemoprevention by O. elatus were apoptosis induction and G2/M cell cycle arrest mediated by a key cell cycle regulator cyclin A.


Subject(s)
Colonic Neoplasms , Oplopanax , Apoptosis , Cell Cycle Checkpoints , Chemoprevention , Cyclin A/pharmacology , Cyclins/pharmacology , Diynes , Fatty Alcohols , Humans , Methylene Chloride/pharmacology , Oplopanax/chemistry , Up-Regulation
6.
J Virol ; 89(21): 10821-31, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26292318

ABSTRACT

UNLABELLED: Gammaherpesviruses (GHVs) carry homologs of cellular genes, including those encoding a viral cyclin that promotes reactivation from latent infection. The viral cyclin has reduced sensitivity to host cyclin-dependent kinase inhibitors in vitro; however, the in vivo significance of this is unclear. Here, we tested the genetic requirement for the viral cyclin in mice that lack the host inhibitors p27(Kip1) and p18(INK4c), two cyclin-dependent kinase inhibitors known to be important in regulating B cell proliferation and differentiation. While the viral cyclin was essential for reactivation in wild-type mice, strikingly, it was dispensable for reactivation in mice lacking p27(Kip1) and p18(INK4c). Further analysis revealed that genetic ablation of only p18(INK4c) alleviated the requirement for the viral cyclin for reactivation from latency. p18(INK4c) regulated reactivation in a dose-dependent manner so that the viral cyclin was dispensable in p18(INK4c) heterozygous mice. Finally, treatment of wild-type cells with the cytokine BAFF, a known attenuator of p18(INK4c) function in B lymphocytes, was also able to bypass the requirement for the viral cyclin in reactivation. These data show that the gammaherpesvirus viral cyclin functions specifically to bypass the cyclin-dependent kinase inhibitor p18(INK4c), revealing an unanticipated specificity between a GHV cyclin and a single cyclin-dependent kinase inhibitor. IMPORTANCE: The gammaherpesviruses (GHVs) cause lifelong infection and can cause chronic inflammatory diseases and cancer, especially in immunosuppressed individuals. Many GHVs encode a conserved viral cyclin that is required for infection and disease. While a common property of the viral cyclins is that they resist inhibition by normal cellular mechanisms, it remains unclear how important it is that the GHVs resist this inhibition. We used a mouse GHV that either contained or lacked a viral cyclin to test whether the viral cyclin lost importance when these inhibitory pathways were removed. These studies revealed that the viral cyclin was required for optimal function in normal mice but that it was no longer required following removal or reduced function of a single cellular inhibitor. These data define a very specific role for the viral cyclin in bypassing one cellular inhibitor and point to new methods to intervene with viral cyclins.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p18/metabolism , Cyclins/metabolism , Gammaherpesvirinae/metabolism , Virus Activation/physiology , Virus Latency/physiology , Animals , B-Cell Activating Factor/pharmacology , Cyclin-Dependent Kinase Inhibitor p18/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p18/deficiency , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Cyclins/pharmacology , DNA Primers/genetics , Flow Cytometry , Gammaherpesvirinae/genetics , Immunoblotting , Mice , Neutralization Tests , Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Virus Activation/drug effects
7.
Am J Physiol Gastrointest Liver Physiol ; 307(11): G1073-87, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25301185

ABSTRACT

Extracellular nucleotides via activation of P2 purinergic receptors influence hepatocyte proliferation and liver regeneration in response to 70% partial hepatectomy (PH). Adult hepatocytes express multiple P2Y (G protein-coupled) and P2X (ligand-gated ion channels) purinergic receptor subtypes. However, the identity of key receptor subtype(s) important for efficient hepatocyte proliferation in regenerating livers remains unknown. To evaluate the impact of P2Y2 purinergic receptor-mediated signaling on hepatocyte proliferation in regenerating livers, wild-type (WT) and P2Y2 purinergic receptor knockout (P2Y2-/-) mice were subjected to 70% PH. Liver tissues were analyzed for activation of early events critical for hepatocyte priming and subsequent cell cycle progression. Our findings suggest that early activation of p42/44 ERK MAPK (5 min), early growth response-1 (Egr-1) and activator protein-1 (AP-1) DNA-binding activity (30 min), and subsequent hepatocyte proliferation (24-72 h) in response to 70% PH were impaired in P2Y2-/- mice. Interestingly, early induction of cytokines (TNF-α, IL-6) and cytokine-mediated signaling (NF-κB, STAT-3) were intact in P2Y2-/- remnant livers, uncovering the importance of cytokine-independent and nucleotide-dependent early priming events critical for subsequent hepatocyte proliferation in regenerating livers. Hepatocytes isolated from the WT and P2Y2-/- mice were treated with ATP or ATPγS for 5-120 min and 12-24 h. Extracellular ATP alone, via activation of P2Y2 purinergic receptors, was sufficient to induce ERK phosphorylation, Egr-1 protein expression, and key cyclins and cell cycle progression of hepatocytes in vitro. Collectively, these findings highlight the functional significance of P2Y2 purinergic receptor activation for efficient hepatocyte priming and proliferation in response to PH.


Subject(s)
Hepatectomy , Hepatocytes/drug effects , Purinergic P2Y Receptor Agonists/pharmacology , Receptors, Purinergic P2Y2/drug effects , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cyclins/pharmacology , Early Growth Response Protein 1/biosynthesis , Early Growth Response Protein 1/genetics , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Purinergic P2Y2/genetics
8.
Reprod Biol ; 24(1): 100827, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38016195

ABSTRACT

Phoenixin-14 (PNX-14) regulates energy metabolism via the G protein-coupled receptor 173 (GPR173); elevated plasma levels have been described in patients with polycystic ovary syndrome. The aims were to investigate the ovarian expression of PNX-14/GPR173 and the in vitro effect of PNX-14 on granulosa cells (Gc) function. Transcript and protein levels of PNX-14/GRP173 were analysed by real-time PCR, western blot and immunohistochemistry in the porcine ovarian follicles at days 2-3, 10-12 and 16-18 of the oestrous. For in vitro experiments, Gc were isolated from follicles at days 10-12 of the oestrous (4-6 mm) and PNX-14 at doses 1-1000 nM was added for 24-72 h to determine Gc proliferation. Cell cycle progression, E2 secretion, expression of proliferating cells nuclear antigen, cyclins, mitogen-activated kinase (MAP3/1; ERK1/2), protein kinase B (AKT) and signal transducer and activator of transcription 3 (STAT3) were studied. The involvement of these kinases in PNX-14 action on Gc proliferation was analysed using pharmacological inhibitors. Levels of GPR173 were increased in the ovarian follicles with oestrous progression, while only PNX-14 protein was the highest at days 10-12 of the oestrous. Immuno-signal of PNX-14 was detected in Gc and theca cells and oocyte, while GPR173 was mostly in theca. Interestingly, PNX-14 stimulated Gc proliferation, E2 secretion, cell cycle progression and cyclins expression and had a modulatory effect on MAP3/1, AKT and STAT3 activation. Our study suggests that PNX-14 could be an important factor for porcine reproduction by influencing ovarian follicle growth through direct action on Gc function.


Subject(s)
Granulosa Cells , Proto-Oncogene Proteins c-akt , Female , Humans , Animals , Swine , Proto-Oncogene Proteins c-akt/metabolism , Ovarian Follicle/metabolism , Ovary , Cyclins/metabolism , Cyclins/pharmacology
9.
Kaohsiung J Med Sci ; 40(2): 175-187, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38010861

ABSTRACT

Intestinal ischemia/reperfusion (I/R) injury is a life-threatening condition with no effective treatment currently available. Curcumin (CCM), a polyphenol compound in Curcuma Longa, reportedly has positive effects against intestinal I/R injury. However, the mechanism underlying the protective effect of CCM against intestinal I/R injury has not been fully clarified. To determine whether the protective effect of CCM was mediated by epigenetic effects on Wnt/ß-catenin signaling, the effect of CCM was examined in vivo and in vitro. An intestinal I/R model was established in Sprague-Dawley (SD) rats with superior mesenteric artery occlusion, and Caco-2 cells were subjected to hypoxia/reoxygenation (H/R) for in vivo simulation of I/R. The results showed that CCM significantly reduced inflammatory, cell apoptosis, and oxidative stress induced by I/R insult in vivo and in vitro. Western blot analysis showed that CCM preconditioning reduced the protein levels of ß-catenin, p-GSK3ß, and cyclin-D1 and increased the protein level of GSK3ß compared with the I/R group. Overexpressing ß-catenin aggravated H/R injury, and knocking down ß-catenin relieved H/R injury by improving intestinal permeability and reducing the cell apoptosis. Moreover, Naked cuticle homolog 2(NKD2) mRNA and protein levels were upregulated in the CCM-pretreated group. 5-aza-2'-deoxycytidine (5-AZA) treatment improved intestinal epithelial barrier impairment induced by H/R. Besides, the protein levels of total ß-catenin, phosphor-ß-catenin and cyclin-D1 were reduced after overexpressing NKD2 in Caco-2 cells following H/R insult. In conclusion, Our study suggests that CCM could attenuate intestinal I/R injury in vitro and in vivo by suppressing the Wnt/ß-catenin signaling pathway via inhibition of NKD2 methylation.


Subject(s)
Curcumin , Reperfusion Injury , Rats , Humans , Animals , Rats, Sprague-Dawley , beta Catenin/genetics , beta Catenin/metabolism , Curcumin/pharmacology , Curcumin/therapeutic use , Wnt Signaling Pathway/genetics , Caco-2 Cells , Glycogen Synthase Kinase 3 beta/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Methylation , Ischemia , Cyclins/metabolism , Cyclins/pharmacology , Apoptosis , Calcium-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism
10.
Expert Opin Ther Targets ; 27(11): 1097-1113, 2023.
Article in English | MEDLINE | ID: mdl-37982244

ABSTRACT

INTRODUCTION: Rheumatoid arthritis (RA) is a chronic inflammatory disease associated with synovial proliferation and bone erosion, which leads to the structural and functional impairment of the joints. Immune cells, together with synoviocytes, induce a pro-inflammatory environment and novel treatment agents target inflammatory cytokines. Psoriasis is a chronic immune-mediated skin disease, and several cytokines are considered as typical mediators in the progression of the disease, including IL-23, IL-22, and IL-17, among others. AREA COVERED: In this review, we try to evaluate whether cyclin-dependent kinases (CDK), enzymes that regulate cell cycle and transcription of various genes, could become novel therapeutic targets in RA and psoriasis. We present the main results of in vitro and in vivo studies, as well as scarce clinical reports. EXPERT OPINION: CDK inhibitors seem promising for treating RA and psoriasis because of their multidirectional effects. CDK inhibitors may affect not only the process of osteoclastogenesis, thereby reducing joint destruction in RA, but also the process of apoptosis of neutrophils and macrophages responsible for the development of inflammation in both RA and psoriasis. However, assessing the efficacy of these drugs in clinical practice requires multi-center, long-term clinical trials evaluating the effectiveness and safety of CDK-blocking therapy in RA and psoriasis.


Subject(s)
Arthritis, Rheumatoid , Psoriasis , Humans , Cyclin-Dependent Kinases/pharmacology , Cyclin-Dependent Kinases/therapeutic use , Arthritis, Rheumatoid/drug therapy , Psoriasis/drug therapy , Cytokines , Cyclins/pharmacology , Cyclins/therapeutic use , Fibroblasts
11.
J Ethnopharmacol ; 312: 116493, 2023 Aug 10.
Article in English | MEDLINE | ID: mdl-37054823

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Lemon myrtle (Backhousia citriodora F.Muell.) leaves, whether fresh or dried, are used traditionally in folk medicine to treat wounds, cancers, skin infections, and other infectious conditions. However, the targets and mechanisms related to anti-cancer effect of lemon myrtle are unavailable. In our study, we found that the essential oil of lemon myrtle (LMEO) showed anti-cancer activity in vitro, and we initially explored its mechanism of action. MATERIALS AND METHODS: We analyzed the chemical compositions of LMEO by GC-MS. We tested the cytotoxicity of LMEO on various cancer cell lines using the MTT assay. Network pharmacology was used also to analyze the targets of LMEO. Moreover, the mechanisms of LMEO were investigated through scratch assay, flow cytometry analysis, and western blot in the HepG2 liver cancer cell line. RESULTS: LMEO showed cytotoxicity on various cancer cell lines with values of IC50 40.90 ± 2.23 (liver cancer HepG2 cell line), 58.60 ± 6.76 (human neuroblastoma SH-SY5Y cell line), 68.91 ± 4.62 (human colon cancer HT-29 cell line) and 57.57 ± 7.61 µg/mL (human non-small cell lung cancer A549 cell line), respectively. The major cytotoxic chemical constituent in LMEO was identified as citrals, which accounted for 74.9% of the content. Network pharmacological analysis suggested that apurinic/apyrimidinic endodeoxyribonuclease 1 (APEX1), androgen receptor (AR), cyclin-dependent kinases 1 (CDK1), nuclear factor erythroid 2-related factor 2 (Nrf-2), fatty acid synthase (FASN), epithelial growth factor receptor (EGFR), estrogen receptor 1 (ERα) and cyclin-dependent kinases 4 (CDK4) are potential cytotoxic targets of LMEO. These targets are closely related to cell migration, cycle and apoptosis. Notley, the p53 protein had the highest confidence to co-associate with the eight common targets, which was further confirmed by scratch assay, flow cytometry analysis, and western blot in the HepG2 liver cancer cell line. LMEO significantly inhibited the migration of HepG2 cells in time-dependent and dose-dependent manner. Moreover, LMEO caused a S-phase blocking on HepG2 cells and promoted apoptosis in the meanwhile. Western blot results indicated that p53 protein, Cyclin A2 and Bax proteins were up-regulated, while Cyclin E1 and Bcl-2 proteins were down-regulated. CONCLUSION: LMEO showed cytotoxicity in various cancer cell lines in vitro. Pharmacological networks showed LMEO to have multi-component and multi-targeting effects that are related to inhibit migration of HepG2 cells, and affect cell cycle S-phase arrest and apoptosis through modulation of p53 protein.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Liver Neoplasms , Lung Neoplasms , Myrtaceae , Myrtus , Neuroblastoma , Oils, Volatile , Humans , Hep G2 Cells , Tumor Suppressor Protein p53/metabolism , Oils, Volatile/chemistry , Carcinoma, Non-Small-Cell Lung/drug therapy , Neuroblastoma/drug therapy , Lung Neoplasms/drug therapy , Cell Cycle , Cell Cycle Checkpoints , Apoptosis , Liver Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Cyclins/metabolism , Cyclins/pharmacology , Cyclins/therapeutic use , Cell Line, Tumor , Cell Proliferation
12.
Mol Brain ; 15(1): 68, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35883152

ABSTRACT

Protein kinases are responsible for protein phosphorylation and are involved in important signal transduction pathways; however, a considerable number of poorly characterized kinases may be involved in neuronal development. Here, we considered cyclin G-associated kinase (GAK) as a candidate regulator of neurite outgrowth and synaptogenesis by examining the effects of the selective GAK inhibitor SGC-GAK-1. SGC-GAK-1 treatment of cultured neurons reduced neurite length and decreased synapse number and phosphorylation of neurofilament 200-kDa subunits relative to the control. In addition, the related kinase inhibitor erlotinib, which has distinct specificity and potency from SGC-GAK-1, had no effect on neurite growth, unlike SGC-GAK-1. These results suggest that GAK may be physiologically involved in normal neuronal development, and that decreased GAK function and the resultant impaired neurite outgrowth and synaptogenesis may be related to neurodevelopmental disorders.


Subject(s)
Cyclic GMP-Dependent Protein Kinases , Cyclins , Cyclic GMP-Dependent Protein Kinases/pharmacology , Cyclin G , Cyclins/pharmacology , Neurites , Neuronal Outgrowth , Protein Kinase Inhibitors/pharmacology , Synapses
13.
Nat Med ; 1(10): 1052-6, 1995 Oct.
Article in English | MEDLINE | ID: mdl-7489362

ABSTRACT

The p21 gene encodes a cyclin-dependent kinase inhibitor that affects cell-cycle progression, but the potential of this gene product to serve as a tumour suppressor in vivo has not been established. In this report, we show that the growth of malignant cells in vitro and in vivo is inhibited by expression of p21. Expression of p21 resulted in an accumulation of cells in G0/G1, altered morphology, and cell differentiation, but apoptosis was not induced. Introduction of p21 with adenoviral vectors into malignant cells completely suppressed their growth in vivo and also reduced the growth of established pre-existing tumours. Gene transfer of p21 may provide a molecular genetic approach to arresting cancer cell growth by committing malignant cells irreversibly to a pathway of terminal differentiation.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/genetics , Cyclins/metabolism , Genes, Tumor Suppressor , Growth Inhibitors/metabolism , 3T3 Cells , Animals , Cell Cycle , Cell Differentiation , Cell Division , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/pharmacology , Gene Transfer Techniques , Humans , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Tumor Cells, Cultured
14.
J Cell Biol ; 113(3): 507-14, 1991 May.
Article in English | MEDLINE | ID: mdl-1826688

ABSTRACT

Functional clam cyclin A and B proteins have been produced using a baculovirus expression system. Both cyclin A and B can induce meiosis I and meiosis II in Xenopus in the absence of protein synthesis. Half-maximal induction occurs at 50 nM for cyclin A and 250 nM for cyclin B. Addition of 25 nM cyclin A to activated Xenopus egg extracts arrested in the cell cycle by treatment with RNase or emetine activates cdc2 kinase to the normal metaphase level and stimulates one oscillatory cell cycle. High levels of cyclin A cause marked hyperactivation of cdc2 kinase and a stable arrest at the metaphase point in the cell cycle. Kinetic studies demonstrate the concentration of cyclin A added does not affect the 10 min lag period required for kinase activation or the timing of maximal activity, but does control the rate of deactivation of cdc2 kinase during exit from mitosis. In addition, exogenous clam cyclin A inhibits the degradation of both A- and B-type endogenous Xenopus cyclins. These results define a system for investigating the biochemistry and regulation of cdc2 kinase activation by cyclin A.


Subject(s)
CDC2 Protein Kinase/metabolism , Cyclins/pharmacology , Animals , Cell Line , Cyclins/metabolism , Cycloheximide/pharmacology , Enzyme Activation , Maturation-Promoting Factor/metabolism , Meiosis/drug effects , Mitosis/drug effects , Oocytes , Protein Biosynthesis , Xenopus laevis
15.
J Cell Biol ; 136(1): 125-35, 1997 Jan 13.
Article in English | MEDLINE | ID: mdl-9008708

ABSTRACT

The replication licensing factor (RLF) is an essential initiation factor that is involved in preventing re-replication of chromosomal DNA in a single cell cycle. In Xenopus egg extracts, it can be separated into two components: RLF-M, a complex of MCM/P1 polypeptides, and RLF-B, which is currently unpurified. In this paper we investigate variations in RLF activity throughout the cell cycle. Total RLF activity is low in metaphase, due to a lack of RLF-B activity and the presence of an RLF inhibitor. RLF-B is rapidly activated on exit from metaphase, and then declines during interphase. The RLF inhibitor present in metaphase extracts is dependent on the activity of cyclin-dependent kinases (Cdks). Affinity depletion of Cdks from metaphase extracts removed the RLF inhibitor, while Cdc2/cyclin B directly inhibited RLF activity. In metaphase extracts treated with the protein kinase inhibitor 6-dimethylaminopurine (6-DMAP), both cyclin B and the RLF inhibitor were stabilized although the extracts morphologically entered interphase. These results are consistent with studies in other organisms that invoke a key role for Cdks in preventing re-replication of DNA in a single cell cycle.


Subject(s)
Cell Cycle Proteins/physiology , Cyclin-Dependent Kinases/physiology , DNA Replication/physiology , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Cell Cycle/physiology , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/pharmacology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/pharmacology , Cycloheximide/pharmacology , DNA Replication/drug effects , Enzyme Inhibitors/pharmacology , Protein Synthesis Inhibitors/pharmacology , Xenopus
16.
J Cell Biol ; 149(6): 1193-206, 2000 Jun 12.
Article in English | MEDLINE | ID: mdl-10851017

ABSTRACT

Many lines of evidence indicate that neoplastic transformation of cells occurs by a multistep process. For neoplastic transformation of normal human cells, they must be first immortalized and then be converted into neoplastic cells. It is well known that the immortalization is a critical step for the neoplastic transformation of cells and that the immortal phenotype is recessive. Thus, we investigated proteins downregulated in immortalized cells by two-dimensional gel electrophoresis. As a result, S100C, a Ca(2+)-binding protein, was dramatically downregulated in immortalized human fibroblasts compared with their normal counterparts. When the cells reached confluence, S100C was phosphorylated on threonine 10. Then the phosphorylated S100C moved to and accumulated in the nuclei of normal cells, whereas in immortalized cells it was not phosphorylated and remained in the cytoplasm. Microinjection of the anti-S100C antibody into normal confluent quiescent cells induced DNA synthesis. Furthermore, when exogenous S100C was compelled to localize in the nuclei of HeLa cells, their DNA synthesis was remarkably inhibited with increase in cyclin-dependent kinase inhibitors such as p16(Ink4a) and p21(Waf1). These data indicate the possible involvement of nuclear S100C in the contact inhibition of cell growth.


Subject(s)
Contact Inhibition , Nuclear Proteins/metabolism , S100 Proteins/metabolism , Amino Acid Sequence , Antibodies, Monoclonal/pharmacology , Carrier Proteins/pharmacology , Cell Division , Cell Line, Transformed , Cell Transformation, Neoplastic , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/pharmacology , DNA/biosynthesis , Electrophoresis, Gel, Two-Dimensional , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique , Humans , Microinjections , Molecular Sequence Data , Neoplasm Proteins/analysis , Phosphopeptides/analysis , Phosphorylation , S100 Proteins/immunology
17.
J Cell Biol ; 137(1): 183-92, 1997 Apr 07.
Article in English | MEDLINE | ID: mdl-9105046

ABSTRACT

Using cell-free extracts made from Xenopus eggs, we show that cdk2-cyclin E and A kinases play an important role in negatively regulating DNA replication. Specifically, we demonstrate that the cdk2 kinase concentration surrounding chromatin in extracts increases 200-fold once the chromatin is assembled into nuclei. Further, we find that if the cdk2-cyclin E or A concentration in egg cytosol is increased 16-fold before the addition of sperm chromatin, the chromatin fails to initiate DNA replication once assembled into nuclei. This demonstrates that cdk2-cyclin E or A can negatively regulate DNA replication. With respect to how this negative regulation occurs, we show that high levels of cdk2-cyclin E do not block the association of the protein complex ORC with sperm chromatin but do prevent association of MCM3, a protein essential for replication. Importantly, we find that MCM3 that is prebound to chromatin does not dissociate when cdk2-cyclin E levels are increased. Taken together our results strongly suggest that during the embryonic cell cycle, the low concentrations of cdk2-cyclin E present in the cytosol after mitosis and before nuclear formation allow proteins essential for potentiating DNA replication to bind to chromatin, and that the high concentration of cdk2-cyclin E within nuclei prevents MCM from reassociating with chromatin after replication. This situation could serve, in part, to limit DNA replication to a single round per cell cycle.


Subject(s)
CDC2-CDC28 Kinases , Cyclin-Dependent Kinases/metabolism , DNA Replication/physiology , Protein Serine-Threonine Kinases/metabolism , S Phase/physiology , Animals , Cell Cycle Proteins/metabolism , Cell Nucleus/physiology , Chromatin/metabolism , Cyclin-Dependent Kinase 2 , Cyclins/metabolism , Cyclins/pharmacology , Cytosol/chemistry , Female , G1 Phase/physiology , G2 Phase/physiology , Male , Oocytes/cytology , Oocytes/enzymology , Protein Binding/physiology , Replication Origin/physiology , Spermatozoa/chemistry , Xenopus , Xenopus Proteins
18.
J Cell Biol ; 149(6): 1179-92, 2000 Jun 12.
Article in English | MEDLINE | ID: mdl-10851016

ABSTRACT

The role of nuclear lamins in DNA replication is unclear. To address this, nuclei were assembled in Xenopus extracts containing AraC, a reversible inhibitor that blocks near the onset of the elongation phase of replication. Dominant-negative lamin mutants lacking their NH(2)-terminal domains were added to assembled nuclei to disrupt lamin organization. This prevented the resumption of DNA replication after the release of the AraC block. This inhibition of replication was not due to gross disruption of nuclear envelope structure and function. The organization of initiation factors was not altered by lamin disruption, and nuclei resumed replication when transferred to extracts treated with CIP, an inhibitor of the cyclin-dependent kinase (cdk) 2-dependent step of initiation. This suggests that alteration of lamin organization does not affect the initiation phase of DNA replication. Instead, we find that disruption of lamin organization inhibited chain elongation in a dose-dependent fashion. Furthermore, the established organization of two elongation factors, proliferating cell nuclear antigen, and replication factor complex, was disrupted by DeltaNLA. These findings demonstrate that lamin organization must be maintained in nuclei for the elongation phase of DNA replication to proceed.


Subject(s)
CDC2-CDC28 Kinases , DNA Replication , Intermediate Filament Proteins , Nuclear Proteins/metabolism , Animals , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/pharmacology , Cytarabine/pharmacology , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique , Humans , Lamin Type B , Lamins , Mutation , Nuclear Envelope/metabolism , Nuclear Localization Signals , Nuclear Proteins/genetics , Oocytes , Peptide Elongation Factors/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Xenopus , Xenopus Proteins
19.
J Cell Biol ; 120(3): 613-24, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8381119

ABSTRACT

A normal consequence of mitosis in eukaryotes is the repression of transcription. Using Xenopus egg extracts shifted to a mitotic state by the addition of purified cyclin, we have for the first time been able to reproduce a mitotic repression of transcription in vitro. Active RNA polymerase III transcription is observed in interphase extracts, but strongly repressed in extracts converted to mitosis. With the topoisomerase II inhibitor VM-26, we demonstrate that this mitotic repression of RNA polymerase III transcription does not require normal chromatin condensation. Similarly; in vitro mitotic repression of transcription does not require the presence of nucleosome structure or involve a general repressive chromatin-binding protein, as inhibition of chromatin formation with saturating amounts of non-specific DNA has no effect on repression. Instead, the mitotic repression of transcription appears to be due to phosphorylation of a component of the transcription machinery by a mitotic protein kinase, either cdc2 kinase and/or a kinase activated by it. Mitotic repression of RNA polymerase III transcription is observed both in complete mitotic cytosol and when a kinase-enriched mitotic fraction is added to a highly simplified 5S RNA transcription reaction. We present evidence that, upon depletion of cdc2 kinase, a secondary protein kinase activity remains and can mediate this in vitro mitotic repression of transcription.


Subject(s)
Mitosis/physiology , Oocytes/physiology , Transcription, Genetic , Animals , CDC2 Protein Kinase/isolation & purification , CDC2 Protein Kinase/metabolism , Cell-Free System , Cyclins/pharmacology , Female , Interphase/physiology , Mitosis/drug effects , Models, Biological , Oocytes/cytology , Protamine Kinase/metabolism , Protein Kinases/isolation & purification , Protein Kinases/metabolism , RNA Polymerase III/metabolism , Templates, Genetic , Teniposide/pharmacology , Topoisomerase II Inhibitors , Transcription Factors/isolation & purification , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Xenopus laevis
20.
Science ; 250(4982): 786-91, 1990 Nov 09.
Article in English | MEDLINE | ID: mdl-2173140

ABSTRACT

The regulation of DNA replication during the eukaryotic cell cycle was studied in a system where cell free replication of simian virus 40 (SV40) DNA was used as a model for chromosome replication. A factor, RF-S, was partially purified from human S phase cells based on its ability to activate DNA replication in extracts from G1 cells. RF-S contained a human homologue of the Schizosaccharomyces pombe p34cdc2 kinase, and this kinase was necessary for RF-S activity. The limiting step in activation of the p34 kinase at the G1 to S transition may be its association with a cyclin since addition of cyclin A to a G1 extract was sufficient to start DNA replication. These observations suggest that the role of p34cdc2 in controlling the start of DNA synthesis has been conserved in evolution.


Subject(s)
CDC2 Protein Kinase/physiology , Cell Cycle , DNA Replication , Simian virus 40/genetics , Virus Replication , Burkitt Lymphoma , CDC2 Protein Kinase/genetics , Cyclins/pharmacology , Humans , Interphase , Phosphorylation , Schizosaccharomyces/enzymology , Simian virus 40/physiology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL