Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 283
Filter
1.
Semin Cancer Biol ; 98: 51-63, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38135020

ABSTRACT

CDK4, along with its regulatory subunit, cyclin D, drives the transition from G1 to S phase, during which DNA replication and metabolic activation occur. In this canonical pathway, CDK4 is essentially a transcriptional regulator that acts through phosphorylation of retinoblastoma protein (RB) and subsequent activation of the transcription factor E2F, ultimately triggering the expression of genes involved in DNA synthesis and cell cycle progression to S phase. In this review, we focus on the newly reported functions of CDK4, which go beyond direct regulation of the cell cycle. In particular, we describe the extranuclear roles of CDK4, including its roles in the regulation of metabolism, cell fate, cell dynamics and the tumor microenvironment. We describe direct phosphorylation targets of CDK4 and decipher how CDK4 influences these physiological processes in the context of cancer.


Subject(s)
Cyclins , Neoplasms , Humans , Cyclins/genetics , Cyclins/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin D/metabolism , Phosphorylation , Cell Cycle/genetics , Neoplasms/genetics , Cell Cycle Proteins/metabolism , Tumor Microenvironment
2.
Organogenesis ; 19(1): 2285836, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-38031805

ABSTRACT

Prostate cancer (PCa) poses a serious burden to men. Interferon-ß (IFN-ß) is implicated in cancer cell growth. This study hence explored the regulation of IFN-ß-modified human umbilical cord mesenchymal stem cell-derived exosomes (hUCMSC-Exos) in PCa cells. In vitro-cultured hUCMSCs were transfected with pcDNA3.1-IFN-ß plasmid or IFN-ß siRNA. hUCMSC-Exos were extracted by ultracentrifugation and identified. PCa cells (PC3 and LNCap) were treated with Exos. Cellular internalization of Exos by cells was detected by uptake assay. Cell proliferation, cycle, and apoptosis were evaluated by CCK-8, EdU staining, and flow cytometry. Levels of cell cycle-related proteins (cyclin D/cyclin E) were determined by Western blot. The effect of IFN-ß-modified hUCMSC-Exos in vivo was analyzed. IFN-ß-modified hUCMSC-Exos (Exooe-IFN-ß or Exosi-IFN-ß) were successfully isolated. IFN-ß was encapsulated in Exos, and PCa cells could uptake Exos. After treating with Exooe-IFN-ß, PCa cell proliferation was impeded, the percentage of cells in the G0/G1 phase, cyclin D/cyclin E levels, and cell apoptotic rate were elevated, while cells treated with Exooe-IFN-ß exhibited contrary trends. IFN-ß-modified hUCMSC-Exos reduced PCa tumor size and weight in vivo. Conjointly, IFN-ß-modified hUCMSC-Exos suppress PCa cell proliferation and facilitate apoptosis.


Subject(s)
Exosomes , Mesenchymal Stem Cells , Prostatic Neoplasms , Male , Humans , Cyclin E/metabolism , Interferon-beta/metabolism , Exosomes/genetics , Exosomes/metabolism , Apoptosis/genetics , Immunologic Factors/metabolism , Prostatic Neoplasms/metabolism , Cell Proliferation , Umbilical Cord/metabolism , Cyclin D/metabolism
3.
Circ Res ; 133(12): 966-988, 2023 12 08.
Article in English | MEDLINE | ID: mdl-37955182

ABSTRACT

BACKGROUND: Pulmonary hypertension (PH) is a chronic vascular disease characterized, among other abnormalities, by hyperproliferative smooth muscle cells and a perturbed cellular redox and metabolic balance. Oxidants induce cell cycle arrest to halt proliferation; however, little is known about the redox-regulated effector proteins that mediate these processes. Here, we report a novel kinase-inhibitory disulfide bond in cyclin D-CDK4 (cyclin-dependent kinase 4) and investigate its role in cell proliferation and PH. METHODS: Oxidative modifications of cyclin D-CDK4 were detected in human pulmonary arterial smooth muscle cells and human pulmonary arterial endothelial cells. Site-directed mutagenesis, tandem mass-spectrometry, cell-based experiments, in vitro kinase activity assays, in silico structural modeling, and a novel redox-dead constitutive knock-in mouse were utilized to investigate the nature and definitively establish the importance of CDK4 cysteine modification in pulmonary vascular cell proliferation. Furthermore, the cyclin D-CDK4 oxidation was assessed in vivo in the pulmonary arteries and isolated human pulmonary arterial smooth muscle cells of patients with pulmonary arterial hypertension and in 3 preclinical models of PH. RESULTS: Cyclin D-CDK4 forms a reversible oxidant-induced heterodimeric disulfide dimer between C7/8 and C135, respectively, in cells in vitro and in pulmonary arteries in vivo to inhibit cyclin D-CDK4 kinase activity, decrease Rb (retinoblastoma) protein phosphorylation, and induce cell cycle arrest. Mutation of CDK4 C135 causes a kinase-impaired phenotype, which decreases cell proliferation rate and alleviates disease phenotype in an experimental mouse PH model, suggesting this cysteine is indispensable for cyclin D-CDK4 kinase activity. Pulmonary arteries and human pulmonary arterial smooth muscle cells from patients with pulmonary arterial hypertension display a decreased level of CDK4 disulfide, consistent with CDK4 being hyperactive in human pulmonary arterial hypertension. Furthermore, auranofin treatment, which induces the cyclin D-CDK4 disulfide, attenuates disease severity in experimental PH models by mitigating pulmonary vascular remodeling. CONCLUSIONS: A novel disulfide bond in cyclin D-CDK4 acts as a rapid switch to inhibit kinase activity and halt cell proliferation. This oxidative modification forms at a critical cysteine residue, which is unique to CDK4, offering the potential for the design of a selective covalent inhibitor predicted to be beneficial in PH.


Subject(s)
Cyclins , Pulmonary Arterial Hypertension , Humans , Mice , Animals , Cyclins/metabolism , Pulmonary Arterial Hypertension/metabolism , Cysteine/metabolism , Endothelial Cells/metabolism , Cell Proliferation , Pulmonary Artery/metabolism , Phosphorylation , Cell Cycle Checkpoints , Cyclin D/metabolism , Cells, Cultured , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism
4.
Zhongguo Zhong Yao Za Zhi ; 48(18): 5049-5055, 2023 Sep.
Article in Chinese | MEDLINE | ID: mdl-37802847

ABSTRACT

This study aimed to explore the effect and mechanism of acetylalkannin from Arnebia euchroma on the proliferation, migration, and invasion of human melanoma A375 cells. A375 cells were divided into a blank group, and low-, medium-, and high-dose acetylalkannin groups(0.5, 1.0, and 2.0 µmol·L~(-1)). The MTT assay was used to detect cell proliferation. Cell scratch and transwell migration assays were used to detect cell migration ability, and the transwell invasion assay was used to detect cell invasion ability. Western blot was used to detect the protein expression of migration and invasion-related N-cadherin, vimentin, matrix metalloproteina-se-9(MMP-9), and Wnt/ß-catenin pathway-related Wnt1, Axin2, glycogen synthase kinase-3ß(GSK-3ß), phosphorylated GSK-3ß(p-GSK-3ß), ß-catenin, cell cycle protein D_1(cyclin D_1), and p21. Real-time fluorescence-based quantitative polymerase chain reaction(real-time PCR) was used to detect the mRNA expression of E-cadherin, matrix metalloproteinase-2(MMP-2), N-cadherin, vimentin, ß-catenin, snail-1, and CD44. MTT results showed that the cell inhibition rates in the acetylalkannin groups significantly increased as compared with that in the blank group(P<0.01). The results of cell scratch and transwell assays showed that compared with the blank group, the acetylalkannin groups showed reduced cell migration and invasion, and migration and invasion rates(P<0.05, P<0.01) and weakened horizontal and vertical migration and invasion abilities. Western blot results showed that compared with the blank group, the high-dose acetylalkannin group showed increased expression of Axin2 protein(P<0.05), and decreased expression of N-cadherin, vimentin, MMP-9, Wnt1, p-GSK-3ß, ß-catenin, cyclin D_1, and p21 proteins(P<0.05, P<0.01). The expression of GSK-3ß protein did not change significantly. PCR results showed that the overall trend of MMP-2, N-cadherin, vimentin, ß-catenin, snail-1, and CD44 mRNA expression was down-regulated(P<0.01), and the expression of E-cadherin mRNA increased(P<0.01). Acetylalkannin can inhibit the proliferation, migration, and invasion of human melanoma A375 cells, and its mechanism of action may be related to the regulation of Wnt/ß-catenin signaling pathway.


Subject(s)
Boraginaceae , Melanoma , Humans , Matrix Metalloproteinase 2/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , beta Catenin/genetics , beta Catenin/metabolism , Vimentin/genetics , Vimentin/metabolism , Matrix Metalloproteinase 9/metabolism , Cell Line, Tumor , Wnt Signaling Pathway , Cadherins/genetics , Melanoma/drug therapy , Melanoma/genetics , Cyclin D/metabolism , Cell Proliferation , Boraginaceae/genetics , RNA, Messenger , Cell Movement
5.
Adv Protein Chem Struct Biol ; 135: 179-201, 2023.
Article in English | MEDLINE | ID: mdl-37061331

ABSTRACT

The recent findings advance our knowledge for the prevention of the premature activation of the major oncogenic pathways including MYC and the cyclin D-cyclin-dependent kinases 4 and 6 (CDK4/6) axis. D-type cyclins are frequently deregulated in human cancer and promote cell division in part through activation of CDK4/6. Therefore, the activation of the cyclin D-CDK4/6 axis stimulates cell proliferation and cancer progression, which represents a unique therapeutic target. However, we have shown that inhibition of CDK4/6 upregulates protein levels of RB1 and CDK6 for acquisition of drug resistance to CDK4/6 inhibitors. Here, we review new progress in the control of cyclin D-dependent cancer cell cycle and proliferation, along with identification of novel E3 ligase for the stability of cyclin D. Cullin4-RING E3 ligase (CRL4)AMBRA1 complex plays a critical role in regulating D-type cyclins through their protein destabilization to control S phase entry and maintain genomic integrity. We also summarize the strategy for inhibition of the cyclin D-associated kinases CDK4/6 and other potential cell cycle regulators for targeting cancer with altered cyclin D expression. We also uncover the function of CK1ɛ as an effective target to potentiate therapeutic efficacy of CDK4/6 inhibitors. Moreover, as the level of PD-L1 is considered in the severe clinical problem in the patients treated with CDK4 inhibitors, we assume that a therapeutic combination using PD-L1 immunotherapy might lower the development of drug resistance and targeting cyclin D will likely inhibit tumor growth and overcome resistance to cyclin D-associated CDK4/6 inhibitors.


Subject(s)
B7-H1 Antigen , Neoplasms , Humans , B7-H1 Antigen/metabolism , Retinoblastoma Protein/metabolism , Phosphorylation , Cyclins/genetics , Cyclin D/metabolism , Signal Transduction , Neoplasms/drug therapy , Carcinogenesis , Cyclin-Dependent Kinase 4/metabolism , Adaptor Proteins, Signal Transducing/metabolism
6.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1008675

ABSTRACT

This study aimed to explore the effect and mechanism of acetylalkannin from Arnebia euchroma on the proliferation, migration, and invasion of human melanoma A375 cells. A375 cells were divided into a blank group, and low-, medium-, and high-dose acetylalkannin groups(0.5, 1.0, and 2.0 μmol·L~(-1)). The MTT assay was used to detect cell proliferation. Cell scratch and transwell migration assays were used to detect cell migration ability, and the transwell invasion assay was used to detect cell invasion ability. Western blot was used to detect the protein expression of migration and invasion-related N-cadherin, vimentin, matrix metalloproteina-se-9(MMP-9), and Wnt/β-catenin pathway-related Wnt1, Axin2, glycogen synthase kinase-3β(GSK-3β), phosphorylated GSK-3β(p-GSK-3β), β-catenin, cell cycle protein D_1(cyclin D_1), and p21. Real-time fluorescence-based quantitative polymerase chain reaction(real-time PCR) was used to detect the mRNA expression of E-cadherin, matrix metalloproteinase-2(MMP-2), N-cadherin, vimentin, β-catenin, snail-1, and CD44. MTT results showed that the cell inhibition rates in the acetylalkannin groups significantly increased as compared with that in the blank group(P<0.01). The results of cell scratch and transwell assays showed that compared with the blank group, the acetylalkannin groups showed reduced cell migration and invasion, and migration and invasion rates(P<0.05, P<0.01) and weakened horizontal and vertical migration and invasion abilities. Western blot results showed that compared with the blank group, the high-dose acetylalkannin group showed increased expression of Axin2 protein(P<0.05), and decreased expression of N-cadherin, vimentin, MMP-9, Wnt1, p-GSK-3β, β-catenin, cyclin D_1, and p21 proteins(P<0.05, P<0.01). The expression of GSK-3β protein did not change significantly. PCR results showed that the overall trend of MMP-2, N-cadherin, vimentin, β-catenin, snail-1, and CD44 mRNA expression was down-regulated(P<0.01), and the expression of E-cadherin mRNA increased(P<0.01). Acetylalkannin can inhibit the proliferation, migration, and invasion of human melanoma A375 cells, and its mechanism of action may be related to the regulation of Wnt/β-catenin signaling pathway.


Subject(s)
Humans , Matrix Metalloproteinase 2/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , beta Catenin/metabolism , Vimentin/metabolism , Matrix Metalloproteinase 9/metabolism , Cell Line, Tumor , Wnt Signaling Pathway , Cadherins/genetics , Melanoma/genetics , Cyclin D/metabolism , Cell Proliferation , Boraginaceae/genetics , RNA, Messenger , Cell Movement
7.
Science ; 378(6622): 845, 2022 11 25.
Article in English | MEDLINE | ID: mdl-36423267
8.
In Vitro Cell Dev Biol Anim ; 58(7): 579-586, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35915338

ABSTRACT

Checkpoint kinase 1 (Chk1) is a protein kinase which preserves the genome integrity, and works as an evolutionally conserved DNA damage response and cell cycle checkpoint. However, the functional roles and regulatory mechanism of Chk1 in mouse granulosa cells (GCs) have not been fully elucidated. In this study, by RNA interfering, Chk1 gene was knocked down in GCs. Knockdown of Chk1 inhibited proliferation and increased apoptosis of GCs (p < 0.05), respectively; in addition, cell cycle of GCs was arrested at S and G2/M phases. Further qRT-PCR results showed that cell cycle factors (Cyclin B1 and Cyclin D 1) and a marker gene of proliferation (PCNA) were downregulated (p < 0.001), while apoptotic factors (p53b, p21, caspase-3, and Bax) were upregulated (p < 0.01), which suggested that knockdown of Chk1 may inhibit proliferation, regulate cell cycle, and promote apoptosis at the transcriptional level in GCs. In vitro studies showed a negative correlation between Chk1 mRNA and miR-16 expression during follicular development. To elucidate the relationship between Chk1 and miR-15a/16, luciferase reporter plasmids were constructed and luciferase assays revealed that both miR-15a and miR-16 could bind to the 3' UTR of Chk1 mRNA, and significantly downregulate the protein level of Chk1 (p < 0.01), while miR-16, not miR-15a, could significantly decrease the mRNA level of Chk1 (p < 0.05). This result indicated that miR-16 directly induced Chk1 mRNA destabilization, while miR-15a regulated Chk1 expression through translational repression. Taken together, this study uncovered the roles of Chk1 in mouse granulosa cells and its regulation by miR-15a and miR-16 through different mechanisms.


Subject(s)
Apoptosis , Checkpoint Kinase 1/metabolism , MicroRNAs , 3' Untranslated Regions , Animals , Apoptosis/genetics , Caspase 3/metabolism , Cell Proliferation/genetics , Checkpoint Kinase 1/genetics , Cyclin B1/metabolism , Cyclin D/metabolism , Female , Granulosa Cells/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , RNA, Messenger/genetics , bcl-2-Associated X Protein/genetics
9.
Oncogene ; 41(30): 3778-3790, 2022 07.
Article in English | MEDLINE | ID: mdl-35761036

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the deadliest cancers. The retinoblastoma protein (RB1), a regulator of cell proliferation, is functionally inactivated in HCC by CYCLIN D/E-mediated phosphorylation. However, the mechanism of RB1-inactivation is unclear because only small percentages of HCCs exhibit amplification of CYCLIN D/E or mutations in the CDK-inhibitory genes. We show that FOXM1, which is overexpressed and critical for HCC, plays essential roles in inactivating RB1 and suppressing RB1-induced senescence of the HCC cells. Mechanistically, FOXM1 binds RB1 and DNMT3B to repress the expression of FOXO1, leading to a decrease in the levels of the CDK-inhibitors, creating an environment for phosphorylation and inactivation of RB1. Consistent with that, inhibition of FOXM1 causes increased expression of FOXO1 with consequent activation of RB1, leading to senescence of the HCC cells, in vitro and in vivo. Also, repression-deficient mutants of FOXM1 induce senescence that is blocked by depletion of RB1 or FOXO1. We provide evidence that human HCCs rely upon this FOXM1-FOXO1 axis for phosphorylation and inactivation of RB1. The observations demonstrate the existence of a new autoregulatory loop of RB1-inactivation in HCC involving a FOXM1-FOXO1 axis that is required for phosphorylation of RB1 and for aggressive progression of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Cellular Senescence , Cyclin D/metabolism , Forkhead Box Protein M1/genetics , Forkhead Box Protein M1/metabolism , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/pathology , Retinoblastoma Binding Proteins/genetics , Retinoblastoma Binding Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
10.
Comput Math Methods Med ; 2022: 1466991, 2022.
Article in English | MEDLINE | ID: mdl-35592685

ABSTRACT

Objective: To explore the mechanism of metformin in treating CCRCC. Methods: Prospective cohort study was conducted. SOD and cyclin D in six CCRCC samples donated by volunteers were detected to compare the degree of oxidative stress injury and the status of cell proliferation. 786-0 CCRCC cells were cultured in vitro with different concentrations of metformin, and MTT assay and Transwell cell migration and wound healing assay were used to detect their proliferation and migration. After culture, SOD and cyclin D in 786-0 CCRCC cells were also detected. Results: In the edge tissue, SOD was lower than in the tumor nest and normal tissue, and cyclin D was highly expressed. In grade II CCRCC, SOD was higher than in grade IV CCRCC, but cyclin D was also highly expressed in grade IV CCRCC. The cell proliferation rate and density of the metformin group were lower than the control group, while in the high-concentration metformin group, it was lower than medium- and low-concentration groups. After culture, the migration of 786-0 cells in the metformin group was significantly lower than that in the control group, the wound healing rate was decreased, and the migration and wound healing rates in the high-concentration metformin group were significantly lower than those in the medium- and low-concentration groups. However, the SOD of the metformin group was higher than the control group, but the cyclin D was lower, while the SOD was higher than medium- and low-concentration groups in the high-concentration group, but the cyclin D was lower after cultured. Conclusion: High-concentration metformin can reduce oxidative stress injury, increase the expression of SOD in CCRCC, and reduce cyclin D in CCRCC to inhibit proliferation and migration, which has optimistic prospects and application value in controlling the progression of CCRCC.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Metformin , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cyclin D/metabolism , Humans , Kidney Neoplasms/metabolism , Metformin/pharmacology , Oxidative Stress , Prospective Studies , Superoxide Dismutase
11.
Pharmacol Res ; 180: 106249, 2022 06.
Article in English | MEDLINE | ID: mdl-35533805

ABSTRACT

Cyclin-dependent kinase 3 (CDK3) is a major player driving retinoblastoma (Rb) phosphorylation during the G0/G1 transition and in the early G1 phase of the cell cycle, preceding the effects of CDK4/cyclin D, CDK6/cyclin D, and CDK2/cyclin E. CDK3 can also directly regulate the activity of E2 factor (E2F) by skipping the role of Rb in late G1, potentially via the phosphorylation of the E2F1 partner DP1. Beyond the cell cycle, CDK3 interacts with various transcription factors involved in cell proliferation, differentiation, and transformation driven by the epidermal growth factor receptor (EGFR)/rat sarcoma virus (Ras) signaling pathway. The expression of CDK3 is extremely low in normal human tissue but upregulated in many cancers, implying a profound role in oncogenesis. Further evaluation of this role has been hampered by the lack of selective pharmacological inhibitors. Herein, we provide a comprehensive overview about the therapeutic potential of targeting CDK3 in cancer.


Subject(s)
Neoplasms , Animals , Cell Cycle , Cyclin D/metabolism , Cyclin-Dependent Kinase 3/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Phosphorylation
12.
Cancer Chemother Pharmacol ; 89(1): 31-40, 2022 01.
Article in English | MEDLINE | ID: mdl-34655298

ABSTRACT

BACKGROUND: Osteosarcoma is a prevalent type of bone tumor in children and adolescents, with limited treatment and poor prognosis. Abemaciclib, an inhibitor of cyclin-dependent kinases 4 and 6 (CDK4/6), is approved for the treatment of advanced breast cancer as single agent therapy and is currently under investigation in clinical trials for the treatment of several solid tumors. METHODS: The efficacy of abemaciclib was determined using osteosarcoma cellular assays and xenograft mouse model. The combination studies were performed based on the Chou-Talalay method. Immunoblotting analysis was performed to determine the underlying mechanisms of abemaciclib in osteosarcoma cell lines. RESULTS: Abemaciclib potently inhibits growth, anchorage-independent colony formation and survival of a panel of osteosarcoma cell lines, with IC50 range from 90 nM to >20 µM. In addition, the combination of abemaciclib and doxorubicin is synergistic and antagonistic in abemaciclib-sensitive (IC50 <1 µM) and abemaciclib-resistant (IC50 >1 µM), respectively. Abemaciclib inhibits tumor formation and growth in a dose-dependent manner without causing significant drug toxicity in mice. The combination of abemaciclib and doxorubicin results in much greater efficacy than doxorubicin alone in inhibiting tumor growth throughout the whole treatment duration. Abemaciclib acts on osteosarcoma via suppressing CDK4/6-Cyclin D-Rb pathway. CONCLUSIONS: Our pre-clinical evidence provides a rationale of initializing clinical trial of investigating the efficacy of abemaciclib in combination with doxorubicin in osteosarcoma patients. Our work also highlights the therapeutic value of CDK4/6 inhibition in osteosarcoma with proper function of Rb.


Subject(s)
Aminopyridines/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzimidazoles/pharmacology , Bone Neoplasms/drug therapy , Osteosarcoma/drug therapy , Aminopyridines/administration & dosage , Animals , Benzimidazoles/administration & dosage , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cyclin D/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Doxorubicin/administration & dosage , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Metabolic Networks and Pathways/drug effects , Mice, SCID , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/pathology , Retinoblastoma Protein/metabolism , Xenograft Model Antitumor Assays
13.
Cells ; 10(11)2021 11 04.
Article in English | MEDLINE | ID: mdl-34831231

ABSTRACT

Resistance to CDK4/6 inhibitors (CDKis) is emerging as a clinical challenge. Identification of the factors contributing to CDKi resistance, with mechanistic insight, is of pivotal significance. Recent studies linked aberrant FGFR signaling to CDKi resistance. However, detailed mechanisms are less clear. Based on control and FGFR1 overexpressing luminal A cell line models, we demonstrated that FGFR1 overexpression rendered the cells resistant to palbociclib. FGFR1 overexpression abolished palbociclib-mediated cell cycle arrest, as well as the attenuated palbociclib-induced inhibition of G1/S transition regulators (pRb, E2F1, and cyclin D3) and factors that promote G2/M transition (cyclin B1, cdc2/CDK1, and cdc25). Importantly, FGFR1-induced palbociclib resistance was associated with promotion of cancer cell stemness and the upregulation of Wnt/ß-catenin signaling. We found that palbociclib may function as an ER agonist in MCF-7/FGFR1 cells. Upregulation of the ER-mediated transcription in MCF-7/FGFR1 cells was associated with ERα phosphorylation and enhanced receptor tyrosine kinase signaling. The combination of palbociclib with FGFR-targeting AZD4547 resulted in remarkable synergistic effects on MCF-7/FGFR1 cells, especially for the inhibition of cancer cell stemness. Our findings of FGFR1-induced palbociclib resistance, promotion of cancer stem cells and associated molecular changes advance our mechanistic understanding of CDKi resistance, which will facilitate the development of strategies targeting CDKi resistance in breast cancer treatment.


Subject(s)
Breast Neoplasms/pathology , Drug Resistance, Neoplasm , Extracellular Signal-Regulated MAP Kinases/metabolism , Neoplastic Stem Cells/metabolism , Piperazines/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Pyridines/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptors, Estrogen/metabolism , Benzamides/pharmacology , Breast Neoplasms/genetics , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Humans , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Neoplasm Invasiveness , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Phosphorylation/drug effects , Pyrazoles/pharmacology , Retinoblastoma Protein/metabolism , Transcription, Genetic/drug effects , Up-Regulation/drug effects , Wnt Signaling Pathway/drug effects
14.
Cells ; 10(9)2021 09 09.
Article in English | MEDLINE | ID: mdl-34572014

ABSTRACT

Despite Glioblastoma (GBM) frequently expressing programmed cell death ligand-1 (PD-L1), treatment with anti-programmed cell death-1 (PD1) has not yielded brilliant results. Intratumor variability of PD-L1 can impact determination accuracy. A previous study on mouse embryonic fibroblasts (MEFs) reported a role for cyclin-D in control of PD-L1 expression. Because tumor-cell growth within a cancer is highly heterogeneous, we looked at whether PD-L1 and its cochaperone FKBP51s were influenced by cell proliferation, using U251 and SF767 GBM-cell-lines. PD-L1 was measured by Western blot, flow cytometry, confocal-microscopy, quantitative PCR (qPCR), CCND1 by qPCR, FKBP51s by Western blot and confocal-microscopy. Chromatin-Immunoprecipitation assay (xChIp) served to assess the DNA-binding of FKBP51 isoforms. In the course of cell culture, PD-L1 appeared to increase concomitantly to cyclin-D on G1/S transition, to decrease during exponential cell growth progressively. We calculated a correlation between CCND1 and PD-L1 gene expression levels. In the temporal window of PD-L1 and CCND1 peak, FKBP51s localized in ER. When cyclin-D declined, FKBP51s went nuclear. XChIp showed that FKBP51s binds CCND1 gene in a closed-chromatin configuration. Our finding suggests that the dynamism of PD-L1 expression in GBM follows cyclin-D fluctuation and raises the hypothesis that FKBP51s might participate in the events that govern cyclin-D oscillation.


Subject(s)
B7-H1 Antigen/metabolism , Brain Neoplasms/metabolism , Cyclin D/metabolism , Glioblastoma/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Fibroblasts/metabolism , Flow Cytometry/methods , Humans
15.
Sci Rep ; 11(1): 14736, 2021 07 19.
Article in English | MEDLINE | ID: mdl-34282211

ABSTRACT

During early G1 phase, Rb is exclusively mono-phosphorylated by cyclin D:Cdk4/6, generating 14 different isoforms with specific binding patterns to E2Fs and other cellular protein targets. While mono-phosphorylated Rb is dispensable for early G1 phase progression, interfering with cyclin D:Cdk4/6 kinase activity prevents G1 phase progression, questioning the role of cyclin D:Cdk4/6 in Rb inactivation. To dissect the molecular functions of cyclin D:Cdk4/6 during cell cycle entry, we generated a single cell reporter for Cdk2 activation, RB inactivation and cell cycle entry by CRISPR/Cas9 tagging endogenous p27 with mCherry. Through single cell tracing of Cdk4i cells, we identified a time-sensitive early G1 phase specific Cdk4/6-dependent phosphorylation gradient that regulates cell cycle entry timing and resides between serum-sensing and cyclin E:Cdk2 activation. To reveal the substrate identity of the Cdk4/6 phosphorylation gradient, we performed whole proteomic and phospho-proteomic mass spectrometry, and identified 147 proteins and 82 phospho-peptides that significantly changed due to Cdk4 inhibition in early G1 phase. In summary, we identified novel (non-Rb) cyclin D:Cdk4/6 substrates that connects early G1 phase functions with cyclin E:Cdk2 activation and Rb inactivation by hyper-phosphorylation.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , G1 Phase/physiology , Cell Division , Cells, Cultured , Cyclin D/metabolism , Cyclin E/metabolism , Humans , Oncogene Proteins/metabolism , Phosphorylation , Proteome/metabolism , Proto-Oncogene Proteins/metabolism , Retinoblastoma Protein/metabolism
16.
Nat Commun ; 12(1): 3356, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099663

ABSTRACT

Since their discovery as drivers of proliferation, cyclin-dependent kinases (CDKs) have been considered therapeutic targets. Small molecule inhibitors of CDK4/6 are used and tested in clinical trials to treat multiple cancer types. Despite their clinical importance, little is known about how CDK4/6 inhibitors affect the stability of CDK4/6 complexes, which bind cyclins and inhibitory proteins such as p21. We develop an assay to monitor CDK complex stability inside the nucleus. Unexpectedly, treatment with CDK4/6 inhibitors-palbociclib, ribociclib, or abemaciclib-immediately dissociates p21 selectively from CDK4 but not CDK6 complexes. This effect mediates indirect inhibition of CDK2 activity by p21 but not p27 redistribution. Our work shows that CDK4/6 inhibitors have two roles: non-catalytic inhibition of CDK2 via p21 displacement from CDK4 complexes, and catalytic inhibition of CDK4/6 independent of p21. By broadening the non-catalytic displacement to p27 and CDK6 containing complexes, next-generation CDK4/6 inhibitors may have improved efficacy and overcome resistance mechanisms.


Subject(s)
Cyclin D/metabolism , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Protein Kinase Inhibitors/pharmacology , Animals , Cell Cycle/drug effects , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , MCF-7 Cells , Mice , Microscopy, Fluorescence , Piperazines/pharmacology , Protein Binding , Pyridines/pharmacology , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism
17.
Int J Mol Sci ; 22(11)2021 May 24.
Article in English | MEDLINE | ID: mdl-34074034

ABSTRACT

In certain diseases of the pancreas, pancreatic stellate cells form an important part of fibrosis and are critical for the development of cancer cells. A hypoxic condition develops within the tumor, to which pancreatic stellate cells adapt and are able to proliferate. The consequence is the growth of the tumor. Melatonin, the product of the pineal gland, is gaining attention as an agent with therapeutic potential against pancreatic cancers. Its actions on tumor cells lead, in general, to a reduction in cell viability and proliferation. However, its effects on pancreatic stellate cells subjected to hypoxia are less known. In this study, we evaluated the actions of pharmacological concentrations of melatonin (1 mM-1 µM) on pancreatic stellate cells subjected to hypoxia. The results show that melatonin induced a decrease in cell viability at the highest concentrations tested. Similarly, the incorporation of BrdU into DNA was diminished by melatonin. The expression of cyclins A and D also was decreased in the presence of melatonin. Upon treatment of cells with melatonin, increases in the expression of major markers of ER stress, namely BIP, phospho-eIF2α and ATF-4, were detected. Modulation of apoptosis was noticed as an increase in caspase-3 activation. In addition, changes in the phosphorylated state of p44/42, p38 and JNK MAPKs were detected in cells treated with melatonin. A slight decrease in the content of α-smooth muscle actin was detected in cells treated with melatonin. Finally, treatment of cells with melatonin decreased the expression of matrix metalloproteinases 2, 3, 9 and 13. Our observations suggest that melatonin, at pharmacological concentrations, diminishes the proliferation of pancreatic stellate cells subjected to hypoxia through modulation of cell cycle, apoptosis and the activation of crucial MAPKs. Cellular responses might involve certain ER stress regulator proteins. In view of the results, melatonin could be taken into consideration as a potential therapeutic agent for pancreatic fibrosis.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Cyclin A/metabolism , Cyclin D/metabolism , Melatonin/pharmacology , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/metabolism , Actins/metabolism , Activating Transcription Factor 4/metabolism , Animals , Bromodeoxyuridine/metabolism , Caspase 3/metabolism , Cell Hypoxia , Cell Survival/drug effects , Cells, Cultured , Endoplasmic Reticulum Stress/drug effects , Eukaryotic Initiation Factor-2/metabolism , Heat-Shock Proteins/metabolism , MAP Kinase Signaling System/drug effects , Matrix Metalloproteinases/metabolism , Rats , Rats, Wistar , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Anticancer Res ; 41(5): 2239-2245, 2021 May.
Article in English | MEDLINE | ID: mdl-33952450

ABSTRACT

BACKGROUND/AIM: This study was designed to investigate the effect of IL-39 on T24 bladder cancer (BC) cell line survival and growth. MATERIALS AND METHODS: In order to assess the direct effect of IL-39 on survival, proliferation, and apoptosis of T24 BC cells, we utilized a clonogenic survival assay, a cell proliferation assay, and caspase-3 activity kits. Potential proliferative and apoptotic molecular mechanisms were evaluated by RT-PCR. RESULTS: Treatment of T24 BC cells with IL-39 resulted in a significant reduction in the percentage of colonies. The anti-tumor effect of IL-39 on T24 bladder cancer cells correlated strongly with a decrease in cyclin E, in combination with an increase in the mRNA levels of Fas. CONCLUSION: IL-39 impedes the growth and survival of T24 BC cells by inhibiting growth and promoting apoptosis. This ability to modulate gene transcription in neoplastic cells shows promise and warrants further research in immunotherapy.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Cyclin E/metabolism , Interleukins/pharmacology , fas Receptor/metabolism , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cyclin D/genetics , Cyclin D/metabolism , Cyclin E/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Reverse Transcriptase Polymerase Chain Reaction , fas Receptor/genetics
19.
Cancer Cell ; 39(6): 759-778, 2021 06 14.
Article in English | MEDLINE | ID: mdl-33891890

ABSTRACT

Abnormal activity of the core cell-cycle machinery is seen in essentially all tumor types and represents a driving force of tumorigenesis. Recent studies revealed that cell-cycle proteins regulate a wide range of cellular functions, in addition to promoting cell division. With the clinical success of CDK4/6 inhibitors, it is becoming increasingly clear that targeting individual cell-cycle components may represent an effective anti-cancer strategy. Here, we discuss the potential of inhibiting different cell-cycle proteins for cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Neoplasms/pathology , Animals , Cell Cycle/drug effects , Cell Cycle/physiology , Cyclin D/genetics , Cyclin D/metabolism , Cyclin E/genetics , Cyclin E/metabolism , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/metabolism , Humans , Mice , Molecular Targeted Therapy/methods
20.
Nature ; 592(7856): 799-803, 2021 04.
Article in English | MEDLINE | ID: mdl-33854232

ABSTRACT

Mammalian development, adult tissue homeostasis and the avoidance of severe diseases including cancer require a properly orchestrated cell cycle, as well as error-free genome maintenance. The key cell-fate decision to replicate the genome is controlled by two major signalling pathways that act in parallel-the MYC pathway and the cyclin D-cyclin-dependent kinase (CDK)-retinoblastoma protein (RB) pathway1,2. Both MYC and the cyclin D-CDK-RB axis are commonly deregulated in cancer, and this is associated with increased genomic instability. The autophagic tumour-suppressor protein AMBRA1 has been linked to the control of cell proliferation, but the underlying molecular mechanisms remain poorly understood. Here we show that AMBRA1 is an upstream master regulator of the transition from G1 to S phase and thereby prevents replication stress. Using a combination of cell and molecular approaches and in vivo models, we reveal that AMBRA1 regulates the abundance of D-type cyclins by mediating their degradation. Furthermore, by controlling the transition from G1 to S phase, AMBRA1 helps to maintain genomic integrity during DNA replication, which counteracts developmental abnormalities and tumour growth. Finally, we identify the CHK1 kinase as a potential therapeutic target in AMBRA1-deficient tumours. These results advance our understanding of the control of replication-phase entry and genomic integrity, and identify the AMBRA1-cyclin D pathway as a crucial cell-cycle-regulatory mechanism that is deeply interconnected with genomic stability in embryonic development and tumorigenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cyclin D/metabolism , Genomic Instability , S Phase , Animals , Cell Line , Cell Proliferation , Checkpoint Kinase 1/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , DNA Replication , Gene Expression Regulation, Developmental , Genes, Tumor Suppressor , Humans , Mice , Mice, Knockout , Synthetic Lethal Mutations
SELECTION OF CITATIONS
SEARCH DETAIL
...