Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Article in English | MEDLINE | ID: mdl-29661866

ABSTRACT

Following antiretroviral therapy, HIV-infected patients show increased circulating levels of the antidiabetic hormone fibroblast growth factor 21 (FGF21). In contrast, the expression of the FGF21-obligatory coreceptor ß-Klotho (KLB) is reduced in target tissues. This situation is comparable to the FGF21 resistance status observed in obesity and type 2 diabetes. Here, we performed the first systematic study of the effects of distinct members of different antiretroviral drug classes on the FGF21/KLB system in human hepatic, adipose, and skeletal muscle cells. Most protease inhibitors and the nonnucleoside reverse transcriptase inhibitor efavirenz induced FGF21 gene expression. Neither nucleoside reverse transcriptase inhibitors nor the viral entry inhibitor maraviroc had any effect. Among the integrase inhibitors, elvitegravir significantly induced FGF21 expression, whereas raltegravir had minor effects only in adipose cells. In human hepatocytes and adipocytes, known target cells of FGF21 action, efavirenz, elvitegravir, and the lopinavir-ritonavir combination exerted inhibitory effects on KLB gene expression. Drug treatments that elicited FGF21 induction/KLB repression were those found to induce endoplasmic reticulum (ER) stress and oxidative stress. Notably, the pharmacological agents thapsigargin and tunicamycin, which induce these stress pathways, mimicked the effects of drug treatments. Moreover, pharmacological inhibitors of either ER or oxidative stress significantly impaired lopinavir-ritonavir-induced regulation of FGF21, but not KLB. In conclusion, the present in vitro screen study identifies the antiretroviral drugs that affect FGF21/KLB expression in human cells. The present results could have important implications for the management of comorbidities resulting from side effects of specific antiretroviral drugs for the treatment of HIV-infected patients.


Subject(s)
Adipose Tissue/metabolism , Anti-Retroviral Agents/pharmacology , Fibroblast Growth Factors/analysis , HIV Infections/drug therapy , Liver/metabolism , Membrane Proteins/analysis , Muscle, Skeletal/metabolism , Alkynes , Benzoxazines/pharmacology , Cyclopropanes , Diabetes Mellitus, Type 2/pathology , Drug Combinations , Endoplasmic Reticulum Stress/drug effects , HIV Integrase Inhibitors/pharmacology , Hep G2 Cells , Humans , Klotho Proteins , Lopinavir/pharmacology , Maraviroc/pharmacology , Obesity/pathology , Oxidative Stress/drug effects , Protease Inhibitors/pharmacology , Quinolones/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Ritonavir/pharmacology , Thapsigargin/pharmacology , Tunicamycin/pharmacology
2.
Mol Oncol ; 17(7): 1228-1245, 2023 07.
Article in English | MEDLINE | ID: mdl-37081792

ABSTRACT

Cyclin-dependent kinases (CDKs), together with their cyclin partners, are the master cell cycle regulators. Remarkably, the cyclin family was extended to include atypical cyclins, characterized by distinctive structural features, but their partner CDKs remain elusive. Here, we conducted a yeast two-hybrid screen to identify new atypical cyclin-CDK complexes. We identified 10 new complexes, including a complex between CDK6 and cyclin I (CCNI), which was found to be active against retinoblastoma protein. CCNI upregulation increased the proliferation of breast cancer cells in vitro and in vivo, with a magnitude similar to that seen upon cyclin D upregulation, an effect that was abrogated by CDK6 silencing or palbociclib treatment. In line with these findings, CCNI downregulation led to a decrease in cell number and a reduction in the percentage of cells reaching S phase. Finally, CCNI upregulation correlated with the high expression of E2F target genes in large panels of cancer cell lines and tissue samples from breast cancer patients. In conclusion, we unveil CCNI as a new player in the pathways that activate CDK6, enriching the wiring of cell cycle control.


Subject(s)
Breast Neoplasms , Cyclin I , Humans , Female , Cyclin I/genetics , Cyclins/genetics , Cyclins/metabolism , Cell Proliferation/genetics , Breast Neoplasms/genetics , Gene Expression , Cell Cycle Proteins/genetics , Cell Cycle , Cyclin-Dependent Kinase 6/genetics
3.
Cell Oncol (Dordr) ; 44(6): 1273-1286, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34604945

ABSTRACT

PURPOSE: Cancer stem cells represent a cancer cell subpopulation that has been found to be associated with metastasis and chemoresistance. Therefore, it is vital to identify mechanisms regulating cancer stemness. Previously, we have shown that the atypical cyclin P (CCNP), also known as CNTD2, is upregulated in lung and colorectal cancers and is associated with a worse clinical prognosis. Given that other cyclins have been implicated in pluripotency regulation, we hypothesized that CCNP may also play a role in cancer stemness. METHODS: Cell line-derived spheroids, ex vivo intestinal organoid cultures and induced-pluripotent stem cells (iPSCs) were used to investigate the role of CCNP in stemness. The effects of CCNP on cancer cell stemness and the expression of pluripotency markers and ATP-binding cassette (ABC) transporters were evaluated using Western blotting and RT-qPCR assays. Cell viability was assessed using a MTT assay. The effects of CCNP on WNT targets were monitored by RNA-seq analysis. Data from publicly available web-based resources were also analyzed. RESULTS: We found that CCNP increases spheroid formation in breast, lung and colorectal cancers, and upregulates the expression of stemness (CD44, CD133) and pluripotency (SOX2, OCT4, NANOG) markers. In addition, we found that CCNP promotes resistance to anticancer drugs and induces the expression of multidrug resistance ABC transporters. Our RNA-seq data indicate that CCNP activates the WNT pathway, and that inhibition of this pathway abrogates the increase in spheroid formation promoted by CCNP. Finally, we found that CCNP knockout decreases OCT4 expression in iPSCs, further supporting the notion that CCNP is involved in stemness regulation. CONCLUSION: Our results reveal CCNP as a novel player in stemness and as a potential therapeutic target in cancer.


Subject(s)
Cyclins/metabolism , Neoplastic Stem Cells/metabolism , Wnt Signaling Pathway , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cyclins/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Neoplastic Stem Cells/pathology , Pluripotent Stem Cells/metabolism , Wnt Signaling Pathway/genetics
4.
Exp Mol Med ; 51(4): 1-17, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30992425

ABSTRACT

CDK16 (also known as PCTAIRE1 or PCTK1) is an atypical member of the cyclin-dependent kinase (CDK) family that forms an active complex with cyclin Y (CCNY). Although both proteins have been recently implicated in cancer pathogenesis, it is still unclear how the CDK16/CCNY complex exerts its biological activity. To understand the CDK16/CCNY network, we used complementary proteomic approaches to identify potential substrates of this complex. We identified several candidates implicating the CDK16/CCNY complex in cytoskeletal dynamics, and we focused on the microtubule-associated protein regulator of cytokinesis (PRC1), an essential protein for cell division that organizes antiparallel microtubules and whose deregulation may drive genomic instability in cancer. Using analog-sensitive (AS) CDK16 generated by CRISPR-Cas9 mutagenesis in 293T cells, we found that specific inhibition of CDK16 induces PRC1 dephosphorylation at Thr481 and delocalization to the nucleus during interphase. The observation that CDK16 inhibition and PRC1 downregulation exhibit epistatic effects on cell viability confirms that these proteins can act through a single pathway. In conclusion, we identified PRC1 as the first substrate of the CDK16/CCNY complex and demonstrated that the proliferative function of CDK16 is mediated by PRC1 phosphorylation. As CDK16 is emerging as a critical node in cancer, our study reveals novel potential therapeutic targets.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinases/metabolism , Cell Cycle Proteins/genetics , Cell Division/genetics , Cell Division/physiology , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Cyclin-Dependent Kinases/genetics , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Phosphorylation , Protein Binding/genetics , Protein Binding/physiology
5.
Sci Rep ; 8(1): 7047, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29728589

ABSTRACT

All-trans-retinoic acid (RA), the active metabolite of vitamin A, can reduce the malignant phenotype in some types of cancer and paradoxically also can promote cancer growth and invasion in others. For instance, it has been reported that RA induces tumor suppression in tumor xenografts of MDA-MB-468 breast cancer cells while increasing tumor growth and metastases in xenografts of MDA-MB-231 breast cancer cells. The signaling pathways involved in the pro-invasive action of retinoic acid remain mostly unknown. We show here that RA activates the pro-invasive axis Src-YAP-Interleukin 6 (Src-YAP-IL6) in triple negative MDA-MB-231 breast cancer cells, yielding to increased invasion of these cells. On the contrary, RA inhibits the Src-YAP-IL6 axis of triple-negative MDA-MB-468 cells, which results in decreased invasion phenotype. In both types of cells, inhibition of the Src-YAP-IL6 axis by the Src inhibitor PP2 drastically reduces migration and invasion. Src inhibition also downregulates the expression of a pro-invasive isoform of VEGFR1 in MDA-MB-231 breast cancer cells. Furthermore, interference of YAP nuclear translocation using the statin cerivastatin reverses the upregulation of Interleukin 6 (IL-6) and the pro-invasive effect of RA on MDA-MB-231 breast cancer cells and also decreases invasion and viability of MDA-MB-468 breast cancer cells. These results altogether suggest that RA induces pro-invasive or anti-invasive actions in two triple-negative breast cancer cell lines due to its ability to activate or inhibit the Src-YAP-IL6 axis in different cancer cells. The pro-invasive effect of RA can be reversed by the statin cerivastatin.


Subject(s)
Interleukin-6/metabolism , Nuclear Proteins/metabolism , Pyridines/pharmacology , Signal Transduction/drug effects , Transcription Factors/metabolism , Tretinoin/pharmacology , Triple Negative Breast Neoplasms/metabolism , src-Family Kinases/metabolism , Biomarkers , Cell Cycle Proteins , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Phosphorylation
6.
Sci Rep ; 8(1): 11797, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30087414

ABSTRACT

Colorectal cancer (CRC) is one of the most common cancers worldwide, with 8-10% of these tumours presenting a BRAF (V600E) mutation. Cyclins are known oncogenes deregulated in many cancers, but the role of the new subfamily of atypical cyclins remains elusive. Here we have performed a systematic analysis of the protein expression levels of eight atypical cyclins in human CRC tumours and several cell lines, and found that CNTD2 is significantly upregulated in CRC tissue compared to the adjacent normal one. CNTD2 overexpression in CRC cell lines increases their proliferation capacity and migration, as well as spheroid formation capacity and anchorage-independent growth. Moreover, CNTD2 increases tumour growth in vivo on xenograft models of CRC with wild-type BRAF. Accordingly, CNTD2 downregulation significantly diminished the proliferation of wild-type BRAF CRC cells, suggesting that CNTD2 may represent a new prognostic factor and a promising drug target in the management of CRC.


Subject(s)
Cell Movement , Cell Proliferation , Colonic Neoplasms/metabolism , Cyclins/metabolism , Mutation, Missense , Proto-Oncogene Proteins B-raf/metabolism , Amino Acid Substitution , Animals , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Cyclins/genetics , Female , Humans , Male , Mice , Mice, Nude , Proto-Oncogene Proteins B-raf/genetics
SELECTION OF CITATIONS
SEARCH DETAIL