Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 399
1.
FASEB J ; 37(5): e22931, 2023 05.
Article En | MEDLINE | ID: mdl-37086099

Premature ovarian failure (POF) is a complication of ovarian dysfunction resulting from the depletion or dysfunction of primordial follicles (PFs) in the ovaries. However, residual follicles that have the potential to be activated are present in POF or aged women. Little is known about the mechanisms by which the remaining dormant PFs in POF patients are activated. Using mass spectrometry, we screened differentially generated peptides extracted from the ovarian cortical tissue biopsies of patients with or without POF, during which we identified PFAP1, a peptide that significantly promoted the activation of PFs in the ovaries of 3 dpp mice in vitro. PFAP1 reversed age-related fertility damage in vivo to a certain extent, promoted estrogen (E2) and anti-mullerian hormone (AMH) production (p < .05), and decreased the levels of follicle-stimulating hormone (FSH) (p < .05). In newborn mouse ovaries, PFAP1 could bind to the protein minichromosome maintenance protein 5 (MCM5) and inhibit its ubiquitination and degradation. In addition, PFAP1 promoted the proliferation of GCs, probably by regulating the function and production of MCM5. In conclusion, PFAP1 could promote the activation of PFs in the ovaries of newborn mice, partially restore the ovarian function of aged mice, and increase the proliferation of primary granulosa cells (GCs) by regulating the function of MCM5. PFAP1 is a promising novel peptide that may be developed into a new therapeutic agent for POF and other ovarian diseases.


Menopause, Premature , Ovarian Diseases , Ovarian Follicle , Peptides , Primary Ovarian Insufficiency , Animals , Female , Mice , Anti-Mullerian Hormone , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Follicle Stimulating Hormone/metabolism , Granulosa Cells/metabolism , Menopause, Premature/metabolism , Ovarian Diseases/drug therapy , Ovarian Diseases/pathology , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Primary Ovarian Insufficiency/metabolism , Peptides/pharmacology
2.
Exp Mol Pathol ; 129: 104850, 2023 02.
Article En | MEDLINE | ID: mdl-36623636

Colorectal cancer (CRC) is a common gastrointestinal tumour with increasing incidence worldwide. However, the underlying molecular mechanism of CRC proliferation is not completely clear. Diversin,as an ankyrin repeat-containing protein, is upregulated in various solid tumours and accelerates cancer progression by promoting cell proliferation and increasing S phase fraction of cells. In this study, 71 CRC samples and corresponding adjacent tissue samples were included. The expression of diversin in tissues was verified via immunohistochemical analysis. The MTS assay and flow cytometry (FCM) was used to measure cell proliferation and cell cycle. Results of immunohistochemical analysis revealed that diversin was highly expressed in human CRC tissues and was significantly associated with tumour differentiation, clinical stage and lymph node metastasis. The analysis based on the CRC data from The Cancer Genome Atlas (TCGA) database showed that a high expression of diversin correlated with the poor prognosis of CRC. Results of the MTS assay indicated that the overexpression of diversin promoted the proliferation of CRC cells, while its downregulation had an inhibitory effect on CRC cell proliferation. FCM analysises presented that diversin increased the flux of the CRC cell cycle from G1 to S and regulated cycle-related proteins, namely, P21, P27, cyclin E, CDK2, cyclin D and CDK4. The results suggest that diversin contributes to CRC proliferation that involves the distribution of the cell cycle. In CRC tissues, the expression of diversin has closely related to the prognosis. The higher the expression levels of diversin, the worse the prognosis. In vitro, diversin could increase the proliferative ability of CRC cells through the G1-S checkpoint and JNK signalling pathway, confirming that diversin contributes to CRC development.


Cell Cycle Proteins , Colorectal Neoplasms , Cytoskeletal Proteins , Humans , Cell Cycle/genetics , Cell Cycle Checkpoints , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Gene Expression Regulation, Neoplastic
3.
Int J Oncol ; 60(3)2022 Mar.
Article En | MEDLINE | ID: mdl-35103287

Oral cancer is a leading cause of cancer­related death worldwide. Current treatment for oral cancer includes surgery, radiotherapy, and chemotherapy; however, their effectiveness is still limited. To identify a new prognostic biomarker and therapeutic target for oral cancer, the Opa interacting protein 5 (OIP5), which plays an essential role in the proper segregation of chromosomes, was examined. Immunohistochemical staining using tissue microarrays indicated that OIP5 was expressed in 120 of 164 (73.2%) oral cancers but was minimally expressed in normal oral tissues. OIP5 expression was significantly associated with poor prognosis in patients with oral cancer. Overexpression of OIP5 enhanced the growth of oral cancer cells, whereas OIP5 knockdown using small interfering RNAs (siRNAs) significantly inhibited cell growth through cell cycle arrest at the G2/M phase. Suppression of OIP5 expression also induced senescence of oral cancer cells. Overall, the findings of the present study suggest that OIP5 may be a candidate prognostic biomarker and therapeutic target in oral cancer.


Cell Cycle Proteins/analysis , Chromosomal Proteins, Non-Histone/analysis , Mouth Neoplasms/drug therapy , Analysis of Variance , Biomarkers, Tumor/analysis , Biomarkers, Tumor/blood , Cell Cycle Proteins/blood , Cell Cycle Proteins/drug effects , Cell Line, Tumor/drug effects , Cell Proliferation/genetics , Chromosomal Proteins, Non-Histone/blood , Chromosomal Proteins, Non-Histone/drug effects , Humans , Mouth Neoplasms/physiopathology
4.
Biomed Pharmacother ; 147: 112652, 2022 Mar.
Article En | MEDLINE | ID: mdl-35065514

Alterations in epigenetic marking, due to changes in expression or activity of epigenetic regulators, may affect cancer development and progression and thus, targeting epigenetic regulators provides potential avenues for cancer treatment. Bromodomain and extra terminal domain (BET) proteins, epigenetic readers recognizing histone acetylation, and Sirtuins (SIRT1-7), histone deacetylases or erasers, affect the chromatin acetylation status, and thus have a vital role in transcriptional regulation of a variety of cancer-related genes. Here, the effects of three BET inhibitors on SIRT expression were screened in a broad set of cancer cell lines to study the potential interplay of these distinct epigenetic factors in gene regulation. We show that BET inhibitors have distinct effects on SIRTs and their target gene expression in cancer cell lines derived from several solid tumour cancers. This functional link may open further avenues for epigenetic combination therapies for different cancers.


Azepines/pharmacology , Benzazepines/pharmacology , Benzodiazepines/pharmacology , Isoxazoles/pharmacology , Proteins/metabolism , Sirtuins/drug effects , Triazoles/pharmacology , Cell Cycle Proteins/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Proteins/drug effects , Transcription Factors/drug effects
5.
J Pharm Pharm Sci ; 25: 69-76, 2022.
Article En | MEDLINE | ID: mdl-35030074

PURPOSE: Among abused substances, methamphetamine is a psychostimulant drug widely used recreationally with public health importance. This study investigated the effect of methamphetamine on proliferation, differentiation, and apoptosis of human adipose tissue stem cells (AdSCs). METHODS: AdSCs were isolated from human abdominal adipose tissue and were characterized for mesenchymal properties and growth kinetics. MTT assay was undertaken to assess methamphetamine toxicity on proliferation and differentiation properties and apoptosis of hAdSCs. RESULTS: Isolated cells were shown to have mesenchymal properties and a population doubling time (PDT) of 40.1 h. Following methamphetamine treatment, expressions of KI-67 and TPX2 as proliferation genes and Col1A1 and PPARg as differentiation genes decreased. Methamphetamine administration increased the expression of Bax and decreased Bcl-2 genes responsible for apoptosis. CONCLUSIONS: Our data suggested when AdSCs were exposed to methamphetamine, it decreased proliferation and differentiation properties of stem cells together with an increase in apoptosis. These findings can be added to the literature, especially when methamphetamine is used recreationally for weight loss purposes.


Adipose Tissue/drug effects , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Mesenchymal Stem Cells/drug effects , Methamphetamine/pharmacology , Adipose Tissue/cytology , Cell Cycle Proteins/drug effects , Genes, bcl-2/drug effects , Humans , Ki-67 Antigen/drug effects , Microtubule-Associated Proteins/drug effects , PPAR gamma/drug effects , bcl-2-Associated X Protein/blood , bcl-2-Associated X Protein/drug effects
6.
Int J Oncol ; 60(2)2022 Feb.
Article En | MEDLINE | ID: mdl-35059737

Low expression levels of the E3 ubiquitin­protein ligase Parkin (PARK2) are exhibited in several cancer entities, including clear cell renal cell carcinoma (ccRCC), and are associated with poor prognosis; however, PARK2 can also function as a tumor suppressor gene. The aim of the present study was to thoroughly investigate the effects of PARK2 overexpression in ccRCC cell lines and to determine its effects on malignancy by conducting functional assays such as cell cycle analysis, apoptosis analysis, migration and invasion assays. Furthermore, liquid chromatography­mass spectrometry was used to decipher potential targets of PARK2 that may influence the behavior of ccRCC tumor cells. In addition, ccRCC tumor tissues from a patient cohort were examined in tissue microarrays to find correlations between different clinical parameters. In the present study, it was demonstrated that the induction of PARK2 resulted in a less aggressive phenotype, as indicated by lower migration and invasion in ccRCC cell lines. Mass spectrometry revealed decreased levels of 29 proteins in cells with PARK2 overexpression, including CDC28 protein kinase regulatory subunit 2 (CKS2), which is highly expressed in numerous types of cancer. The link between the function of PARK2 as an E3 ubiquitin ligase and the low expression levels of CKS2 was investigated by mutating the catalytic domain of the PARK2 gene, and it was found that the effect of decreased migration was abolished in 786­O and RCC­MH ccRCC cell lines. CKS2 silencing decreased migratory ability of the cells. Furthermore, it was revealed that high CKS2 levels are associated with high tumor grading in patient samples and lower patient survival. In conclusion, the results from the present study indicated that PARK2 may signal via CKS2 to affect tumor behavior. In consequence, CKS2 may be a biomarker in ccRCC and may also serve as potential target for ccRCC therapy.


CDC2-CDC28 Kinases/drug effects , Carcinoma, Renal Cell/drug therapy , Cell Cycle Proteins/drug effects , Ubiquitin-Protein Ligases/pharmacology , CDC2-CDC28 Kinases/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor/metabolism , Cell Proliferation/drug effects , Cell Proliferation/genetics , Disease Progression , Humans , Ubiquitin-Protein Ligases/administration & dosage , Ubiquitin-Protein Ligases/metabolism
7.
Clin Transl Med ; 12(1): e660, 2022 01.
Article En | MEDLINE | ID: mdl-35075807

OBJECTIVE: To explore the therapeutic potential and the underlying mechanism of metformin, an adenosine monophosphate-activated kinase (AMPK) activator, in ocular melanoma. METHODS: CCK8, transwell, and colony formation assays were performed to detect the proliferation and migration ability of ocular melanoma cells. A mouse orthotopic xenograft model was built to detect ocular tumor growth in vivo. Western blot, immunofluorescence, and electron microscopy were adopted to evaluate the autophagy levels of ocular melanoma cells, and high-throughput proteomics and CUT & Tag assays were performed to analyze the candidate for autophagy alteration. RESULTS: Here, we revealed for the first time that a relatively low dose of metformin induced significant tumorspecific inhibition of the proliferation and migration of ocular melanoma cells both in vitro and in vivo. Intriguingly, we found that metformin significantly attenuated autophagic influx in ocular melanoma cells. Through high-throughput proteomics analysis, we revealed that optineurin (OPTN), which is a key candidate for autophagosome formation and maturation, was significantly downregulated after metformin treatment. Moreover, excessive OPTN expression was associated with an unfavorable prognosis of patients. Most importantly, we found that a histone deacetylase, SIRT1, was significantly upregulated after AMPK activation, resulting in histone deacetylation in the OPTN promoter. CONCLUSIONS: Overall, we revealed for the first time that metformin significantly inhibited the progression of ocular melanoma, and verified that metformin acted as an autophagy inhibitor through histone deacetylation of OPTN. This study provides novel insights into metformin - guided suppression of ocular melanoma and the potential mechanism underlying the dual role of metformin in autophagy regulation.


Autophagy/drug effects , Cell Cycle Proteins/drug effects , Histone Demethylases/drug effects , Melanoma/drug therapy , Membrane Transport Proteins/drug effects , Metformin/agonists , Animals , Cell Cycle Proteins/metabolism , Disease Models, Animal , Eye/drug effects , Eye/metabolism , Melanoma/metabolism , Membrane Transport Proteins/metabolism , Metformin/therapeutic use , Mice , Neoplasms/drug therapy , Neoplasms/metabolism
8.
Clin Transl Med ; 12(1): e718, 2022 01.
Article En | MEDLINE | ID: mdl-35083874

BACKGROUND: Chemoradiotherapy-induced PD-L1 upregulation leads to therapeutic resistance and treatment failure. The PD-1/PD-L1 blocking antibodies sensitize cancers to chemoradiotherapy by blocking extracellular PD-1 and PD-L1 binding without affecting the oncogenic function of intracellular PD-L1. Reversing the chemoradiation-induced PD-L1 expression could provide a new strategy to achieve a greater anti-tumour effect of chemoradiotherapy. Here, we aimed to identify candidate small molecular inhibitors that might boost the anti-tumour immunity of chemoradiotherapy by decreasing treatment-induced PD-L1 expression in non-small cell lung cancer (NSCLC). METHODS: A drug array was used to recognize compounds that can suppress the cisplatin-induced and radiation-induced PD-L1 expression in NSCLC via the flow cytometry-based assay. We examined whether and how targeting bromodomain containing 4 (BRD4) inhibits chemoradiation-induced PD-L1 expression and evaluated the effect of BRD4 inhibition and chemoradiation combination in vivo. RESULTS: BRD4 inhibitors JQ1 and ARV-771 were identified as the most promising drugs both in the cisplatin and radiation screening projects in two NSCLC cell lines. Targeting BRD4 was supposed to block chemoradiotherapy inducible PD-L1 expression by disrupting the recruitment of BRD4-IRF1 complex to PD-L1 promoter. A positive correlation between BRD4 and PD-L1 expression was observed in human NSCLC tissues. Moreover, BRD4 inhibition synergized with chemoradiotherapy and PD-1 blockade to show a robust anti-tumour immunity dependent on CD8+ T cell through limiting chemoradiation-induced tumour cell surface PD-L1 upregulation in vivo. Notably, the BRD4-targeted combinatory treatments did not show increased toxicities. CONCLUSION: The data showed that BRD4-targeted therapy synergized with chemoradiotherapy and anti-PD-1 antibody by boosting anti-tumour immunity in NSCLC.


Carcinoma, Non-Small-Cell Lung/drug therapy , Chemoradiotherapy/standards , Signal Transduction/genetics , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/genetics , Chemoradiotherapy/methods , Chemoradiotherapy/statistics & numerical data , Disease Models, Animal , Gene Expression/drug effects , Gene Expression/genetics , Interferon Regulatory Factor-1/drug effects , Interferon Regulatory Factor-1/genetics , Mice , Signal Transduction/drug effects , Transcription Factors/drug effects , Transcription Factors/genetics
9.
Oncol Rep ; 47(1)2022 Jan.
Article En | MEDLINE | ID: mdl-34751409

Promoter methylation represents one of the major epigenetic mechanisms responsible for the regulation of gene expression. Hypomethylating drugs are currently approved for the treatment of myelodysplastic syndromes and acute myeloid leukemia, and some studies have recently been carried out on diffuse large B cell lymphoma (DLBCL). DLBCL is a type of Non­Hodgkin lymphoma. The aim of the present study was to assess the role of DNA methyltransferase (DNMT)1 in mediating the epigenetic regulation of some key targets previously emerged as hypermethylated in Non­Hodgkin lymphoma. Reverse transcription­quantitative PCR, genome­wide arrays and methylation­specific PCR were used to determine the level of methylation of specific targets. Gene silencing, gene expression and immunoblotting were used to investigate the role of DNMT1 and DNMT3a in lymphoma cells. The present study showed that lymphoma cell lines displayed a completely different methylation profile on selected targets compared with primary B lymphocytes and peripheral blood mononuclear cells. 5'­aza­cytidine (5AZA) and 5'­aza­2­deoxycitidine (decitabine) exerted their activity through, at least in part, mechanisms independent of DNMT1 downregulation. Despite a global hypomethylating effect of 5AZA and decitabine, DNMT1 was not found to be necessary to maintain the hypermethylation of Krüppel­like factor 4 (KLF4), death associated protein 1 (DAPK1) and spastic paraplegia 20 (SPG20). SPG20 was found to be a completely methylated target in all the tested cell lines, but not in peripheral blood mononuclear cells, suggesting its association with malignancy. The highest methylation was clustered upstream of the transcription starting site in a panel of 28 DLBCL cell lines and the results were unaffected by the silencing of DNMT1 expression. These data demonstrated the epigenetic regulation of SPG20 in lymphoid cells and identified a number of novel markers associated with lymphomas that deserve further investigation.


Cell Cycle Proteins/genetics , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Death-Associated Protein Kinases/genetics , Decitabine/pharmacology , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/genetics , Antimetabolites, Antineoplastic/pharmacology , Cell Cycle Proteins/drug effects , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferase 1/drug effects , DNA Methylation/drug effects , DNA Methylation/genetics , Death-Associated Protein Kinases/drug effects , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Kruppel-Like Factor 4/drug effects , Kruppel-Like Factor 4/genetics
10.
Cancer Lett ; 528: 31-44, 2022 03 01.
Article En | MEDLINE | ID: mdl-34942306

The CDK4/6 inhibitor, palbociclib has recently entered clinic-trial stage for breast cancer treatment. However, translating its efficacy to other solid tumors has been challenging, especially for aggressive solid tumors. We found that the effect of palbociclib as a single agent was limited due to primary and acquired resistance in multiple ovarian cancer (OC) models. Among these, patient-derived organoid and xenograft models are two most representative models of drug responsiveness in patients with OC. In preclinical models, this study demonstrated that activated MAPK/PI3K-AKT pathway and cell cycle-related proteins induced the resistance to palbociclib, which was overcome by the addition of the bromodomain protein 4 (BRD4) inhibitor AZD5153. Moreover, this study revealed that AZD5153 and palbociclib had a synergistic lethal effect on inducing the cell cycle arrest and increasing apoptosis, even in RB-deficient cell lines. Based on these results, it is anticipated that this class of drugs, including AZD5153, which inhibit the cell cycle-related protein and MAPK/PI3K-AKT pathway, will exhibit synergistic effects with palbociclib in OC.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Cycle Proteins/drug effects , Heterocyclic Compounds, 2-Ring/therapeutic use , Mitogen-Activated Protein Kinase Kinases/metabolism , Ovarian Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Piperazines/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/therapeutic use , Pyridazines/therapeutic use , Pyridines/therapeutic use , Animals , Antineoplastic Agents , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Female , Heterocyclic Compounds, 2-Ring/pharmacology , Humans , Mice , Piperazines/pharmacology , Pyrazoles/pharmacology , Pyridazines/pharmacology , Pyridines/pharmacology , Xenograft Model Antitumor Assays
11.
Molecules ; 26(19)2021 Sep 24.
Article En | MEDLINE | ID: mdl-34641328

This study aims to enhance efficacy and reduce toxicity of the combination treatment of a drug and curcumin (Cur) on leukemic stem cell and leukemic cell lines, including KG-1a and KG-1 (FLT3+ LSCs), EoL-1 (FLT3+ LCs), and U937 (FLT3- LCs). The cytotoxicity of co-treatments of doxorubicin (Dox) or idarubicin (Ida) at concentrations of the IC10-IC80 values and each concentration of Cur at the IC20, IC30, IC40, and IC50 values (conditions 1, 2, 3, and 4) was determined by MTT assays. Dox-Cur increased cytotoxicity in leukemic cells. Dox-Cur co-treatment showed additive and synergistic effects in several conditions. The effect of this co-treatment on FLT3 expression in KG-1a, KG-1, and EoL-1 cells was examined by Western blotting. Dox-Cur decreased FLT3 protein levels and total cell numbers in all the cell lines in a dose-dependent manner. In summary, this study exhibits a novel report of Dox-Cur co-treatment in both enhancing cytotoxicity of Dox and inhibiting cell proliferation via FLT3 protein expression in leukemia stem cells and leukemic cells. This is the option of leukemia treatment with reducing side effects of chemotherapeutic drugs to leukemia patients.


Curcumin/pharmacology , Doxorubicin/pharmacology , Idarubicin/pharmacology , Leukemia, Myeloid, Acute/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Antigens, Neoplasm/drug effects , Antigens, Neoplasm/metabolism , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Curcuma/chemistry , Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Leukemia, Myeloid, Acute/drug therapy , Rhizome/chemistry
12.
Theranostics ; 11(19): 9180-9197, 2021.
Article En | MEDLINE | ID: mdl-34646365

Cell cycle regulators are frequently altered in Triple-Negative Breast Cancer (TNBC). Emerging agents targeting these signals offer the possibility to design new combinatorial therapies. However, preclinical models that recapitulate TNBC primary resistance and heterogeneity are essential to evaluate the potency of these combined treatments. Methods: Bioinformatic processing of human breast cancer datasets was used to analyse correlations between expression levels of cell cycle regulators and patient survival outcome. The MMTV-R26Met mouse model of TNBC resistance and heterogeneity was employed to analyse expression and targeting vulnerability of cell cycle regulators in the presence of BCL-XL blockage. Robustness of outcomes and selectivity was further explored using a panel of human breast cancer cells. Orthotopic studies in nude mice were applied for preclinical evaluation of efficacy and toxicity. Alterations of protein expression, phosphorylation, and/or cellular localisation were analysed by western blots, reverse phase protein array, and immunocytochemistry. Bioinformatics was performed to highlight drug's mechanisms of action. Results: We report that high expression levels of the BCL2L1 gene encoding BCL-XL and of specific cell cycle regulators correlate with poor survival outcomes of TNBC patients. Blockage of BCL-XL confers vulnerability to drugs targeting CDK1/2/4, but not FOXM1, CDK4/6, Aurora A and Aurora B, to all MMTV-R26Met and human TNBC cell lines tested. Combined blockage of BCL-XL and CDK1/2/4 interfered with tumour growth in vivo. Mechanistically, we show that, co-targeting of BCL-XL and CDK1/2/4 synergistically inhibited cell viability by combinatorial depletion of survival and RTK/AKT signals, and concomitantly restoring FOXO3a tumour suppression actions. This was accompanied by an accumulation of DNA damage and consequently apoptosis. Conclusions: Our studies illustrate the possibility to exploit the vulnerability of TNBC cells to CDK1/2/4 inhibition by targeting BCL-XL. Moreover, they underline that specificity matters in targeting cell cycle regulators for combinatorial anticancer therapies.


Triple Negative Breast Neoplasms/metabolism , bcl-X Protein/metabolism , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Computational Biology , Cyclin-Dependent Kinases/metabolism , DNA Damage/drug effects , Databases, Genetic , Disease Models, Animal , Female , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Mice, Nude , Phosphorylation , Signal Transduction/drug effects , Transcriptome/genetics , Triple Negative Breast Neoplasms/drug therapy , Xenograft Model Antitumor Assays , bcl-X Protein/genetics
13.
Cells ; 10(10)2021 09 23.
Article En | MEDLINE | ID: mdl-34685500

Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive disease with a dismal prognosis. Here, we show how an inhibition of de novo dNTP synthesis by the ribonucleotide reductase (RNR) inhibitor hydroxyurea and an inhibition of epigenetic modifiers of the histone deacetylase (HDAC) family affect short-term cultured primary murine PDAC cells. We used clinically relevant doses of hydroxyurea and the class 1 HDAC inhibitor entinostat. We analyzed the cells by flow cytometry and immunoblot. Regarding the induction of apoptosis and DNA replication stress, hydroxyurea and the novel RNR inhibitor COH29 are superior to the topoisomerase-1 inhibitor irinotecan which is used to treat PDAC. Entinostat promotes the induction of DNA replication stress by hydroxyurea. This is associated with an increase in the PP2A subunit PR130/PPP2R3A and a reduction of the ribonucleotide reductase subunit RRM2 and the DNA repair protein RAD51. We further show that class 1 HDAC activity promotes the hydroxyurea-induced activation of the checkpoint kinase ataxia-telangiectasia mutated (ATM). Unlike in other cell systems, ATM is pro-apoptotic in hydroxyurea-treated murine PDAC cells. These data reveal novel insights into a cytotoxic, ATM-regulated, and HDAC-dependent replication stress program in PDAC cells.


Ataxia Telangiectasia/drug therapy , Enzyme Inhibitors/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/mortality , Animals , Antineoplastic Agents/pharmacology , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , DNA Damage/drug effects , DNA Replication/drug effects , Mice , Pancreatic Neoplasms
14.
BMC Cancer ; 21(1): 1061, 2021 Sep 26.
Article En | MEDLINE | ID: mdl-34565342

BACKGROUND: Neuroblastoma (NB) patients with MYCN amplification or overexpression respond poorly to current therapies and exhibit extremely poor clinical outcomes. PI3K-mTOR signaling-driven deregulation of protein synthesis is very common in NB and various other cancers that promote MYCN stabilization. In addition, both the MYCN and mTOR signaling axes can directly regulate a common translation pathway that leads to increased protein synthesis and cell proliferation. However, a strategy of concurrently targeting MYCN and mTOR signaling in NB remains unexplored. This study aimed to investigate the therapeutic potential of targeting dysregulated protein synthesis pathways by inhibiting the MYCN and mTOR pathways together in NB. METHODS: Using small molecule/pharmacologic approaches, we evaluated the effects of combined inhibition of MYCN transcription and mTOR signaling on NB cell growth/survival and associated molecular mechanism(s) in NB cell lines. We used two well-established BET (bromodomain extra-terminal) protein inhibitors (JQ1, OTX-015), and a clinically relevant mTOR inhibitor, temsirolimus, to target MYCN transcription and mTOR signaling, respectively. The single agent and combined efficacies of these inhibitors on NB cell growth, apoptosis, cell cycle and neurospheres were assessed using MTT, Annexin-V, propidium-iodide staining and sphere assays, respectively. Effects of inhibitors on global protein synthesis were quantified using a fluorescence-based (FamAzide)-based protein synthesis assay. Further, we investigated the specificities of these inhibitors in targeting the associated pathways/molecules using western blot analyses. RESULTS: Co-treatment of JQ1 or OTX-015 with temsirolimus synergistically suppressed NB cell growth/survival by inducing G1 cell cycle arrest and apoptosis with greatest efficacy in MYCN-amplified NB cells. Mechanistically, the co-treatment of JQ1 or OTX-015 with temsirolimus significantly downregulated the expression levels of phosphorylated 4EBP1/p70-S6K/eIF4E (mTOR components) and BRD4 (BET protein)/MYCN proteins. Further, this combination significantly inhibited global protein synthesis, compared to single agents. Our findings also demonstrated that both JQ1 and temsirolimus chemosensitized NB cells when tested in combination with cisplatin chemotherapy. CONCLUSIONS: Together, our findings demonstrate synergistic efficacy of JQ1 or OTX-015 and temsirolimus against MYCN-driven NB, by dual-inhibition of MYCN (targeting transcription) and mTOR (targeting translation). Additional preclinical evaluation is warranted to determine the clinical utility of targeted therapy for high-risk NB patients.


Acetanilides/pharmacology , Azepines/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacology , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , Neuroblastoma/drug therapy , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/antagonists & inhibitors , Triazoles/pharmacology , Adaptor Proteins, Signal Transducing/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Down-Regulation , Drug Synergism , Eukaryotic Initiation Factor-4E/drug effects , Eukaryotic Initiation Factor-4E/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Ribosomal Protein S6 Kinases, 70-kDa/drug effects , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , Sirolimus/pharmacology , Spheroids, Cellular/drug effects , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/drug effects , Transcription Factors/metabolism
15.
Int Heart J ; 62(5): 1112-1123, 2021 Sep 30.
Article En | MEDLINE | ID: mdl-34544967

Doxorubicin (DOX) is a widely used anticancer drug, but its cardiotoxicity largely limits its clinical utilization. Circular RNA spindle and kinetochore-associated protein 3 (circ-SKA3) were found to be differentially expressed in heart failure patients. In this study, we investigated the role and mechanism of circ-SKA3 in DOX-induced cardiotoxicity.The quantitative real-time polymerase chain reaction and western blot assays were applied to measure the expression of circ-SKA3, microRNA (miR) -1303, and toll-like receptor 4 (TLR4). The viability and apoptosis of AC16 cells were analyzed using cell counting kit-8, flow cytometry, and western blot assays. The interaction between miR-1303 and circ-SKA3 or TLR4 was verified using dual-luciferase reporter and RNA immunoprecipitation assays. Exosomes were collected from culture media by the use of commercial kits and then qualified by transmission electron microscopy.The expression of circ-SKA3 and TLR4 was increased, whereas miR-1303 expression was decreased in DOX-treated AC16 cells. DOX treatment promoted cell apoptosis and inhibited cell viability in AC16 cells in vitro, which was partially reversed by circ-SKA3 knockdown, TLR4 silencing, or miR-1303 overexpression. Mechanistically, circ-SKA3 served as a sponge for miR-1303 to upregulate TLR4, which was confirmed to be a target of miR-1303. Additionally, circ-SKA3 contributed to DOX-induced cardiotoxicity through the miR-1303/TLR4 axis. Further studies suggested that circ-SKA3 was overexpressed in exosomes extracted from DOX-mediated AC16 cells, which could be internalized by surrounding untreated AC16 cells.Circ-SKA3 enhanced DOX-induced toxicity in AC16 cells through the miR-1303/TLR4 axis. Extracellular circ-SKA3 was packaged into exosomes, and exosomal circ-SKA3 could function as a mediator in intercellular communication between AC16 cells.


Cell Cycle Proteins/genetics , Doxorubicin/toxicity , Microtubule-Associated Proteins/genetics , Myocytes, Cardiac/drug effects , Topoisomerase II Inhibitors/toxicity , Apoptosis/drug effects , Cardiotoxicity/genetics , Cell Cycle Proteins/drug effects , Cell Survival/drug effects , Exosomes/genetics , Heart Failure/genetics , Humans , MicroRNAs/genetics , Microscopy, Electron, Transmission/methods , Microtubule-Associated Proteins/drug effects , Myocytes, Cardiac/pathology , RNA, Circular/genetics , Toll-Like Receptor 4/drug effects , Toll-Like Receptor 4/genetics , Transfection/methods , Up-Regulation
16.
Am J Physiol Heart Circ Physiol ; 321(2): H259-H274, 2021 08 01.
Article En | MEDLINE | ID: mdl-34085839

Underlying molecular mechanisms for the development of diabetic cardiomyopathy remain to be determined. Long-term exposure to hyperglycemia causes oxidative stress, which leads to cardiomyocyte dysfunction. Previous studies established the importance of thioredoxin-interacting protein (Txnip) in cellular redox homeostasis and glucose metabolism. Txnip is a highly glucose-responsive molecule that interacts with the catalytic center of reduced thioredoxin and inhibits the antioxidant function of thioredoxin. Here, we show that the molecular interaction between Txnip and thioredoxin plays a pivotal role in the regulation of redox balance in the diabetic myocardium. High glucose increased Txnip expression, decreased thioredoxin activities, and caused oxidative stress in cells. The Txnip-thioredoxin complex was detected in cells with overexpressing wild-type Txnip but not Txnip cysteine 247 to serine (C247S) mutant that disrupts the intermolecular disulfide bridge. Then, diabetes was induced in cardiomyocyte-specific Txnip C247S knock-in mice and their littermate control animals by injections of streptozotocin (STZ). Prolonged hyperglycemia upregulated myocardial Txnip expression in both genotypes. The absence of Txnip's inhibition of thioredoxin in Txnip C247S mutant hearts promoted mitochondrial antioxidative capacities in cardiomyocytes, thereby protecting the heart from oxidative damage by diabetes. Stress hemodynamic analysis uncovered that Txnip C247S knock-in hearts have a greater left ventricular contractile reserve than wild-type hearts under STZ-induced diabetic conditions. These results provide novel evidence that Txnip serves as a regulator of hyperglycemia-induced cardiomyocyte toxicities through direct inhibition of thioredoxin and identify the single cysteine residue in Txnip as a therapeutic target for diabetic injuries.NEW & NORTEWORTHY Thioredoxin-interacting protein (Txnip) has been of great interest as a molecular mechanism to mediate diabetic organ damage. Here, we provide novel evidence that a single mutation of Txnip confers a defense mechanism against myocardial oxidative stress in streptozotocin-induced diabetic mice. The results demonstrate the importance of Txnip as a cysteine-containing redox protein that regulates antioxidant thioredoxin via disulfide bond-switching mechanism and identify the cysteine in Txnip as a therapeutic target for diabetic cardiomyopathy.


Carrier Proteins/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetic Cardiomyopathies/metabolism , Myocytes, Cardiac/metabolism , Oxidative Stress/genetics , Thioredoxins/metabolism , Ventricular Function, Left/genetics , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Line , Gene Knock-In Techniques , Glucose/pharmacology , HEK293 Cells , Humans , Isolated Heart Preparation , Mice , Mutation , Myocytes, Cardiac/drug effects , Rats , Thioredoxins/genetics
17.
World Neurosurg ; 153: e46-e58, 2021 09.
Article En | MEDLINE | ID: mdl-34129971

BACKGROUND: Glioma is a common primary intracranial tumor, with high infiltration and aggression. Propofol (Pro) is associated with growth and metastasis in glioma. Meanwhile, circular RNA non-SMC condensin I complex subunit G (circNCAPG; hsa_circ_0007244) has been reported to be upregulated in glioma. This study explored the role and mechanism of circNCAPG in Pro-induced glioma progression. METHODS: Cell viability was determined by cell counting kit-8 assay. Levels of circNCAPG, microRNA-200a-3p (miR-200a-3p), and member RAS oncogene family (RAB5A) were detected by real-time quantitative polymerase chain reaction. Colony number, apoptosis, migration, and invasion were analyzed by colony formation, flow cytometry, wound healing, and transwell assays. Matrix metallopeptidase 2, matrix metallopeptidase 9, and RAB5A protein levels were detected by Western blot assay. The binding relationship between miR-200a-3p and circNCAPG or RAB5A was predicted by starBase 2.0 and then verified by a dual-luciferase reporter and RNA immunoprecipitation assays. The biological roles of circNCAPG and Pro on glioma tumor growth were examined by the xenograft tumor model in vivo. RESULTS: Expression of circNCAPG and RAB5A was upregulated, and miR-200a-3p was decreased in glioma tissues and cells, while their expression presented an opposite trend in Pro-treated glioma cells. Moreover, circNCAPG overexpression could abolish Pro-mediated proliferation, apoptosis, migration, and invasion in glioma cells in vitro. Mechanically, circNCAPG could regulate RAB5A expression by sponging miR-200a-3p. Pro blocked glioma tumor growth in vivo by modulating circNCAPG. CONCLUSIONS: Pro could inhibit glioma cell growth and metastasis through the circNCAPG/miR-200a-3p/RAB5A axis, providing a promising therapeutic strategy for glioma treatment.


Anesthetics, Intravenous/pharmacology , Brain Neoplasms/genetics , Cell Cycle Proteins/drug effects , Glioma/genetics , MicroRNAs/drug effects , Propofol/pharmacology , rab5 GTP-Binding Proteins/drug effects , Adult , Apoptosis/drug effects , Apoptosis/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Female , Glioma/metabolism , Glioma/pathology , Humans , Male , Matrix Metalloproteinase 2/drug effects , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/drug effects , Matrix Metalloproteinase 9/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Neoplasm Invasiveness , Neoplasm Transplantation , RNA, Circular , Real-Time Polymerase Chain Reaction , Tumor Stem Cell Assay , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism
18.
Biomolecules ; 11(5)2021 05 18.
Article En | MEDLINE | ID: mdl-34069817

Unrestrained proliferation is a common feature of malignant neoplasms. Targeting the cell cycle is a therapeutic strategy to prevent unlimited cell division. Recently developed rationales for these selective inhibitors can be subdivided into two categories with antithetical functionality. One applies a "brake" to the cell cycle to halt cell proliferation, such as with inhibitors of cell cycle kinases. The other "accelerates" the cell cycle to initiate replication/mitotic catastrophe, such as with inhibitors of cell cycle checkpoint kinases. The fate of cell cycle progression or arrest is tightly regulated by the presence of tolerable or excessive DNA damage, respectively. This suggests that there is compatibility between inhibitors of DNA repair kinases, such as PARP inhibitors, and inhibitors of cell cycle checkpoint kinases. In the present review, we explore alterations to the cell cycle that are concomitant with altered DNA damage repair machinery in unfavorable neuroblastomas, with respect to their unique genomic and molecular features. We highlight the vulnerabilities of these alterations that are attributable to the features of each. Based on the assessment, we offer possible therapeutic approaches for personalized medicine, which are seemingly antithetical, but both are promising strategies for targeting the altered cell cycle in unfavorable neuroblastomas.


Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Neuroblastoma/metabolism , Antineoplastic Agents/therapeutic use , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , DNA Repair/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neuroblastoma/drug therapy
19.
Neurochem Int ; 147: 105047, 2021 07.
Article En | MEDLINE | ID: mdl-33872680

Mammalian nuclear distribution genes encode proteins with essential roles in neuronal migration and brain formation during embryogenesis. The implication of human nuclear distribution genes, namely nudC and NDE1 (Nuclear Distribution Element 1)/NDEL1 (Nuclear Distribution Element-Like 1), in psychiatric disorders including schizophrenia and bipolar disorder, has been recently described. The partial loss of NDEL1 expression results in neuronal migration defects, while ndel1 null knockout (KO) leads to early embryonic lethality in mice. On the other hand, loss-of-function of the orthologs of nuclear distribution element genes (nud) in Caenorhabditis elegans renders viable worms and influences behavioral endophenotypes associated with dopaminergic and serotoninergic pathways. In the present work, we evaluated the role of nud genes in monoamine levels at baseline and after the treatment with typical or atypical antipsychotics. Dopamine, serotonin and octopamine levels were significantly lower in homozygous loss-of-function mutant worms KO for nud genes compared with wild-type (WT) C. elegans at baseline. While treatment with antipsychotics determined significant differences in monoamine levels in WT, the nud KO mutant worms appear to respond differently to the treatment. According to the best of our knowledge, we are the first to report the influence of nud genes in the monoamine levels changes in response to antipsychotic drugs, ultimately placing the nuclear distribution genes family at the cornerstone of pathways involved in the modulation of monoamines in response to different classes of antipsychotic drugs.


Antipsychotic Agents/pharmacology , Biogenic Monoamines/metabolism , Brain/drug effects , Carrier Proteins/drug effects , Mutation/drug effects , Animals , Brain/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/drug effects , Caenorhabditis elegans Proteins/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Microtubule-Associated Proteins/drug effects , Microtubule-Associated Proteins/metabolism
20.
Clin Transl Oncol ; 23(9): 1847-1856, 2021 Sep.
Article En | MEDLINE | ID: mdl-33821368

BACKGROUND: Hepatocellular carcinoma is one of the most common malignancies and leading cancer-associated deaths worldwide. Ozone has been proposed as a promising therapeutic agent in the treatment of various disorders. PURPOSE: The purpose of this paper is to assess the potential anticancer effects of the ozone on liver cancer cells. METHOD: The liver cancer cell line of bel7402 and SMMC7721 was used in this study. Proliferation was evaluated using the CCK-8 and the colony formation assay. Wond healing assay and transwell assay without Matrigel were used to evaluate their migration ability. Flow cytometry was used for cell cycle analysis and reactive oxygen species (ROS) determination. Glutathione detection kit was used for measurement of glutathione level. Protein expression was estimated by western blot analysis. RESULTS: Ozone treatment inhibited liver cancer cell proliferation, colony formation. Ozone induced G2/M phase cell cycle arrest, which could be elucidated by the change of protein levels of p53, p21, Cyclin D1, cyclin B1, cdc2, and CDK4. We also found that ozone treatment inhibited migration ability by inhibiting EMT-relating protein. Ozone also induced ROS accumulation and decreased glutathione level decreased, which contributed to the inactivation of the PI3K/AKT/NF-κB pathway. Finally, we found that pre-treatment of liver cancer cells with N-acetylcysteine resisted ozone-induced effects. CONCLUSIONS: Ozone restrains the proliferation and migration potential and EMT process of liver cancer cells via ROS accumulation and PI3K/AKT/NF-κB suppression.


Carcinoma, Hepatocellular/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Liver Neoplasms/metabolism , Ozone/pharmacology , Reactive Oxygen Species/metabolism , Carcinoma, Hepatocellular/pathology , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Survival , G2 Phase Cell Cycle Checkpoints/drug effects , Glutathione/metabolism , Humans , Liver Neoplasms/pathology , M Phase Cell Cycle Checkpoints/drug effects , NF-kappa B/drug effects , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Tumor Stem Cell Assay
...