Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Microvasc Res ; 142: 104348, 2022 07.
Article in English | MEDLINE | ID: mdl-35245516

ABSTRACT

BACKGROUND: Retinoblastoma protein (Rb) supports vasoprotective E2F Transcription Factor 1 (E2f1)/Dihydrofolate Reductase (Dhfr) pathway activity in endothelial cells. Cyclin I (Ccni) promotes Cyclin-Dependent Kinase-5 (Cdk5)-mediated Rb phosphorylation. Therefore, we hypothesized that endothelial Ccni may regulate cardiovascular homeostasis, vessel remodeling, and abdominal aortic aneurysm (AAA) formation. METHODS: Aortic CCNI mRNA expression was analyzed in the Gene Expression Omnibus (GEO) GSE57691 cohort consisting of AAA patients (n = 39) and healthy controls (n = 10). We employed wild-type (WT) mice and endothelial Ccni knockout (Ccnifl/flTie2-Cre) mice to conduct in vivo and ex vivo experimentation using an Angiotensin (Ang) II hypertension model and a CaCl2 AAA model. Mice were assessed for Rb/E2f1/Dhfr signaling, biopterin (i.e., biopterin [B], dihydrobiopterin [BH2], and tetrahydrobiopterin [BH4]) production, cardiovascular homeostasis, vessel remodeling, and AAA formation. RESULTS: Aortic CCNI mRNA expression was downregulated in AAA patients. Both Ang II- and CaCl2-induced WT mice showed aortic Ccni upregulation coupled with vasculoprotective upregulation of Rb/E2f1/Dhfr signaling and biopterins. Endothelial Ccni knockout downregulated medial Rb/E2f1/Dhfr signaling and biopterins in Ang II-induced hypertensive mice, which exacerbated eNos uncoupling and H2O2 production. Endothelial Ccni knockout impaired in vivo hemodynamic responses and endothelium-dependent vasodilatation in ex vivo mesenteric arteries in response to Ang II. Endothelial Ccni knockout exacerbated mesenteric artery remodeling and AAA risk in response to Ang II and CaCl2. CONCLUSIONS: Endothelial Ccni acts as a critical negative regulator of eNos uncoupling-mediated ROS generation and thereby reduces vulnerability to hypertension-induced vascular remodeling and AAA development in mice.


Subject(s)
Angiotensin II , Aortic Aneurysm, Abdominal , Hypertension , Vascular Remodeling , Angiotensin II/pharmacology , Animals , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/prevention & control , Biopterins/metabolism , Calcium Chloride/metabolism , Cyclin I/metabolism , Cysteine-Rich Protein 61/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Endothelium/metabolism , Humans , Hydrogen Peroxide/metabolism , Hypertension/genetics , Hypertension/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/metabolism , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism
2.
J Cell Mol Med ; 25(4): 1896-1910, 2021 02.
Article in English | MEDLINE | ID: mdl-33465281

ABSTRACT

Extracellular vesicles (Evs) participate in the development of rheumatoid arthritis (RA), but the mechanisms remain unclear. This study aimed to determine the mechanism by which microRNA-34a (miR-34a) contained in bone marrow mesenchymal stem cell (BM-MSC)-derived Evs functions in RA fibroblast-like synoviocytes (RA-FLSs). BM-MSC-derived Evs and an Evs inhibitor were extracted. A rat model of RA was established. miR-34a gain- and loss-of-function experiments were performed, and the inflammation in rat synovial fluid and tissues was detected. The role of miR-34a in RA-FLSs was also measured in vitro. The target gene of miR-34a was predicted using the online software TargetScan and identified using a dual-luciferase reporter gene assay, and the activation of the ATM/ATR/p53 signalling pathway was assessed. BM-MSC-derived Evs mainly elevated miR-34a expression, which reduced RA inflammation in vivo and inhibited RA-FLS proliferation and resistance to apoptosis in vitro, while inhibited miR-34a expression enhanced RA development. In addition, miR-34a could target cyclin I to activate the ATM/ATR/p53 signalling pathway, thus inhibiting abnormal RA-FLS growth and RA inflammation. Our study showed that miR-34a contained in BM-MSC-derived Evs could reduce RA inflammation by inhibiting the cyclin I/ATM/ATR/p53 signalling pathway.


Subject(s)
Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Extracellular Vesicles/metabolism , Gene Expression Regulation , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Signal Transduction , Animals , Apoptosis/genetics , Arthritis, Rheumatoid/pathology , Biomarkers , Biopsy , Cells, Cultured , Cyclin I/metabolism , Male , MicroRNAs/metabolism , Rats , Synoviocytes/metabolism , Synoviocytes/pathology , Tumor Suppressor Protein p53/metabolism
3.
Am J Physiol Cell Physiol ; 308(4): C339-47, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25500740

ABSTRACT

The atypical cyclin-dependent kinase 5 (Cdk5) serves an array of different functions in cell biology. Among these are axonal guidance, regulation of intercellular contacts, cell differentiation, and prosurvival signaling. The variance of these functions suggests that Cdk5 activation comes to pass in different cellular compartments. The kinase activity, half-life, and substrate specificity of Cdk5 largely depend on specific activators, such as p25, p35, p39, and cyclin I. We hypothesized that the subcellular distribution of Cdk5 activators also determines the localization of the Cdk5 protein and sets the stage for targeted kinase activity within distinct cellular compartments to suit the varying roles of Cdk5. Cdk5 localization was analyzed in murine kidney and brain slices of wild-type and cyclin I- and/or p35-null mice by immunohistochemistry and in cultured mouse podocytes using immunofluorescence labeling, as well as cell fractionation experiments. The predominance of cyclin I mediates the nuclear localization of Cdk5, whereas the predominance of p35 results in a membranous localization of Cdk5. These findings were further substantiated by overexpression of cyclin I and p35 with altered targeting characteristics in human embryonic kidney 293T cells. These studies reveal that the subcellular localization of Cdk5 is determined by its specific activators. This results in the directed Cdk5 kinase activity in specific cellular compartments dependent on the activator present and allows Cdk5 to serve multiple independent roles.


Subject(s)
Cyclin I/metabolism , Cyclin-Dependent Kinase 5/metabolism , Phosphotransferases/metabolism , Podocytes/enzymology , Animals , Cell Membrane/enzymology , Cell Nucleus/enzymology , Cyclin I/deficiency , Cyclin I/genetics , Endoplasmic Reticulum/enzymology , Enzyme Activation , HEK293 Cells , Humans , Mice, Knockout , Phosphotransferases/deficiency , Phosphotransferases/genetics , Protein Transport , Purkinje Cells/enzymology , Transfection
4.
Proteins ; 82(11): 3117-31, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25136968

ABSTRACT

A-to-I RNA editing has been recently shown to be a widespread phenomenon with millions of sites spread in the human transcriptome. However, only few are known to be located in coding sequences and modify the amino acid sequence of the protein product. Here, we used high-throughput data, variant prediction tools, and protein structural information in order to find structural and functional preferences for coding RNA editing. We show that RNA editing has a unique pattern of amino acid changes characterized by enriched stop-to-tryptophan changes, positive-to-neutral and neutral-to-positive charge changes. RNA editing tends to have stronger structural effect than equivalent A-to-G SNPs but weaker effect than random A-to-G mutagenesis events. Sites edited at low level tend to be located at conserved positions with stronger predicted deleterious effect on proteins comparing to sites edited at high frequencies. Lowly edited sites tend to destabilize the protein structure and affect amino acids with larger number of intra-molecular contacts. Still, some highly edited sites are predicted also to prominently affect structure and tend to be located at critical positions of the protein matrix and are likely to be functionally important. Using our pipeline, we identify and discuss several novel putative functional coding changing editing sites in the genes COPA (I164V), GIPC1 (T62A), ZN358 (K382R), and CCNI (R75G).


Subject(s)
Biological Evolution , Proteins/chemistry , Proteins/genetics , RNA Editing , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Amino Acids/genetics , Cyclin I/chemistry , Cyclin I/genetics , Databases, Protein , Humans , Kv1.1 Potassium Channel/chemistry , Kv1.1 Potassium Channel/genetics , Mutagenesis , Polymorphism, Single Nucleotide , Protein Interaction Domains and Motifs , Receptors, AMPA/chemistry , Receptors, AMPA/genetics , Tryptophan
5.
Avian Dis ; 58(3): 468-73, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25518444

ABSTRACT

Tibial dyschondroplasia (TD) is a skeletal disease characterized by the disruption ofendochondral bone formation in avian species. The aim of this study was to determine the expression of Matrilin-3 and Cyclin-I genes in chicken growth plate during impairment and recovery from TD. Gene expressions were analyzed by polymerase chain reaction, and proteins by immunohistochemistry and in situ hybridizations. Expression of genes encoding Matrilin-3 and Cyclin-I were diminished with parallel decrease in proteins during TD, with fewer signs of cartilage cell differentiation. In contrast, there was an increase in mRNA expressions and protein levels of both genes during the recovery phase. These findings suggest that the Matrilin-3 and Cyclin-I genes also play a role in chondrocyte differentiation during the impairment and recovery of growth plate in TD.


Subject(s)
Avian Proteins/genetics , Chickens/genetics , Cyclin I/genetics , Growth Plate/metabolism , Matrilin Proteins/genetics , Osteochondrodysplasias/veterinary , Poultry Diseases/genetics , Animals , Avian Proteins/metabolism , Cell Differentiation , Chickens/metabolism , Chondrocytes/cytology , Chondrocytes/metabolism , Cyclin I/metabolism , Matrilin Proteins/metabolism , Osteochondrodysplasias/genetics , Osteochondrodysplasias/metabolism , Poultry Diseases/metabolism , Recovery of Function , Tibia/metabolism
6.
Biomol Biomed ; 24(2): 323-336, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-37540586

ABSTRACT

Globally, pancreatic cancer is recognized as one of the deadliest malignancies that lacks effective targeted therapies. This study aims to explore the role of cyclin I-like protein (CCNI2), a homolog of cyclin I (CCNI), in the progression of pancreatic cancer, thereby providing a theoretical basis for its treatment. Firstly, the expression of CCNI2 in pancreatic cancer tissues was determined through immunohistochemical staining. The biological role of CCNI2 in pancreatic cancer cells was further assessed using both in vitro and in vivo loss/gain-of-function assays. Our data revealed that CCNI2 expression was abnormally elevated in pancreatic cancer, and clinically, increased CCNI2 expression generally correlated with reduced overall survival. Functionally, CCNI2 contributed to the malignant progression of pancreatic cancer by promoting the proliferation and migration of tumor cells. Consistently, in vivo experiments verified that CCNI2 knockdown impaired the tumorigenic ability of pancreatic cancer cells. Moreover, the addition of phosphatidylinositol 3-kinase (PI3K) inhibitors could partially reverse the promoting effect of CCNI2 on the malignant phenotypes of pancreatic cancer cells. CCNI2 promoted pancreatic cancer through PI3K/protein kinase B (AKT) signaling pathway, indicating its potential as a prognostic marker and therapeutic target for pancreatic cancer.


Subject(s)
Pancreatic Neoplasms , Proto-Oncogene Proteins c-akt , Humans , Proto-Oncogene Proteins c-akt/genetics , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Cyclin I/metabolism , Cell Proliferation/genetics , Signal Transduction , Pancreatic Neoplasms/genetics
7.
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
8.
Diabetes ; 72(1): 85-96, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36201618

ABSTRACT

A recent discovery effort resulted in identification of novel splice variant and secretory granule antigens within the HLA class I peptidome of human islets and documentation of their recognition by CD8+ T cells from peripheral blood and human islets. In the current study, we applied a systematic discovery process to identify novel CD4+ T cell epitopes derived from these candidate antigens. We predicted 145 potential epitopes spanning unique splice junctions and within conventional secretory granule antigens and measured their in vitro binding to DRB1*04:01. We generated HLA class II tetramers for the 35 peptides with detectable binding and used these to assess immunogenicity and isolate T cell clones. Tetramers corresponding to peptides with verified immunogenicity were then used to label T cells specific for these putative epitopes in peripheral blood. T cells that recognize distinct epitopes derived from a cyclin I splice variant, neuroendocrine convertase 2, and urocortin-3 were detected at frequencies that were similar to those of an immunodominant proinsulin epitope. Cells specific for these novel epitopes predominantly exhibited a Th1-like surface phenotype. Among the three epitopes, responses to the cyclin I peptide exhibited a distinct memory profile. Responses to neuroendocrine convertase 2 were detected among pancreatic infiltrating T cells. These results further establish the contribution of unconventional antigens to the loss of tolerance in autoimmune diabetes.


Subject(s)
CD4-Positive T-Lymphocytes , Diabetes Mellitus, Type 1 , Humans , Cyclin I/metabolism , Diabetes Mellitus, Type 1/metabolism , Epitopes, T-Lymphocyte , Peptides/metabolism , Secretory Vesicles , Alternative Splicing
9.
Am J Physiol Renal Physiol ; 302(9): F1161-71, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22262481

ABSTRACT

Cyclin-dependent kinase (Cdk)-5 is activated by both cyclin I and the noncyclin activator p35 in terminally differentiated cells such as kidney podocytes and neurons. Cyclin I and p35 are restricted to podocytes in the kidney, and each limit podocyte apoptosis by activating Cdk5. To determine whether both activators are necessary, or whether they serve backup roles, a double cyclin I-p35 null mouse was generated. Experimental glomerular disease characterized by podocyte apoptosis was then induced by administering an anti-podocyte antibody. The results showed that under nonstressed conditions double mutants had normal kidney structure and function and were indistinguishable from wild-type, cyclin I(-/-), or p35(-/-) mice. In contrast, when stressed with disease, podocyte apoptosis increased fourfold compared with diseased cyclin I(-/-) or p35(-/-) mice. This resulted in a more pronounced decrease in podocyte number, proteinuria, and glomerulosclerosis. Under normal states and nephritic states, levels for the prosurvival protein Bcl-2 were lower in double cyclin I(-/-) p35(-/-) mice than the other mice. Similarly, levels of Bcl-xL, another prosurvival member, were lower in normal and nephritic double cyclin I(-/-) p35(-/-) mice but similar to single-cyclin I(-/-) mice. Moreover, levels of ERK1/2 and MEK1/2 activation were lower in nephritic double cyclin I(-/-) p35(-/-) mice but similar to single-cyclin I(-/-) mice. The results demonstrate that the activators of Cdk5, p35, and cyclin I are not required for normal kidney function. However, they play pivotal coordinated roles in maintaining podocyte survival during stress states in disease.


Subject(s)
Apoptosis/physiology , Cyclin I/metabolism , Cyclin-Dependent Kinase 5/metabolism , Phosphotransferases/metabolism , Podocytes/metabolism , Podocytes/pathology , Animals , Antibodies, Anti-Idiotypic/adverse effects , Cell Survival/physiology , Cyclin I/deficiency , Cyclin I/genetics , Disease Models, Animal , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Kidney Diseases/pathology , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphotransferases/deficiency , Phosphotransferases/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-X Protein/metabolism
10.
Gynecol Oncol ; 127(1): 217-22, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22772062

ABSTRACT

OBJECTIVE: Ovarian cancer is the most lethal of all gynecologic malignancies. It is characterized by the spread of intraperitoneal tumors, accumulation of ascites, and formation of tumor blood vessels. Cyclin I has been linked with angiogenesis-related proteins, like vascular endothelial growth factor (VEGF) and VEGF receptor 2 (VEGFR-2), in human breast cancer. We examined whether an association exists between expression of cyclin I, VEGFR-2, clinicopathologic parameters and survival of patients with epithelial ovarian cancer (EOC). METHODS: Cyclin I and VEGFR-2 expressions were analyzed by immunohistochemistry in 55 human primary EOC tissue specimens. RESULTS: Cyclin I immunoreactivity was significantly correlated with VEGFR-2 (R=0.4587, P=0.0004), and immunolabeling of cyclin I and VEGFR-2 significantly correlated with cancer cells' proliferative activity evaluated using cyclin A labeling index as a marker (R=0.3107, P=0.0209 and R=0.4183, P=0.0015, respectively). VEGFR-2 immunostaining was significantly higher in advanced, poorly differentiated, and suboptimally resected EOCs compared to their counterparts (P<0.05). Finally, higher VEGFR-2 expression was significantly associated with shorter disease-free survival (P=0.0437). CONCLUSIONS: Our results indicate that elevated expression of cyclin I and VEGFR-2 is likely to provide a proliferative advantage to the EOC cells, and that cyclin I may be linked with angiogenesis in EOC. Higher expression of VEGFR-2 is associated with more advanced disease. Further investigation of cyclin I in ovarian cancer is needed to evaluate if cyclin I may become a novel target for an anticancer therapy.


Subject(s)
Cyclin I/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Adult , Aged , Aged, 80 and over , Carcinoma, Ovarian Epithelial , Cell Growth Processes/physiology , Cyclin I/biosynthesis , Female , Humans , Immunohistochemistry , Middle Aged , Neoplasms, Glandular and Epithelial/blood supply , Neoplasms, Glandular and Epithelial/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/pathology , Vascular Endothelial Growth Factor Receptor-2/biosynthesis , Young Adult
11.
ESC Heart Fail ; 9(2): 1370-1379, 2022 04.
Article in English | MEDLINE | ID: mdl-35128826

ABSTRACT

AIMS: Heart failure (HF) is a chronic heart disease with a high incidence and mortality. Due to the regulatory complexity of gene coexpression networks, the underlying hub genes regulation in HF remain incompletely appreciated. We aimed to explore potential key modules and genes for HF using weighted gene coexpression network analysis (WGCNA). METHODS AND RESULTS: The expression profiles by high throughput sequencing of heart tissues samples from HF and non-HF samples were obtained from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) between HF and non-HF samples were firstly identified. Then, a coexpression network was constructed to identify key modules and potential hub genes. The biological functions of potential hub genes were analysed by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses. Finally, a protein-protein interaction (PPI) network was constructed using the STRING online tool. A total of 135 DEGs (133 up-regulated and 2 down-regulated DEGs) between HF and non-HF samples were identified in the GSE135055 and GSE123976 datasets. Moreover, a total of 38 modules were screened based on WGCNA in the GSE135055 dataset, and six potential hub genes (UCK2, ASB1, CCNI, CUX1, IRX6, and STX16) were screened from the key module by setting the gene significance over 0.2 and the module membership over 0.8. Furthermore, 78 potential hub genes were obtained by taking the intersection of the 135 DEGs and all genes in the key module, and enrichment analysis revealed that they were mainly involved in the MAPK and PI3K-AKT signalling pathways. Finally, in a PPI network constructed with the 78 potential hub genes, CUX1 and ASB1 were identified as hub genes in HF because they were also identified as potential hub genes in the WGCNA. CONCLUSIONS: To the best of our knowledge, our study is the first to employ WGCNA to identify the key module and hub genes for HF. Our study identified a module and two genes that might play important roles in HF, which may provide potential biomarkers for the diagnosis of HF and improve our knowledge of the molecular mechanisms underlying HF.


Subject(s)
Heart Failure , Phosphatidylinositol 3-Kinases , Biomarkers/metabolism , Cyclin I , Gene Expression Profiling/methods , Gene Regulatory Networks , Heart Failure/genetics , Homeodomain Proteins , Humans , Transcription Factors/genetics
12.
Cancer Med ; 10(6): 1913-1924, 2021 03.
Article in English | MEDLINE | ID: mdl-33620152

ABSTRACT

Colorectal cancer (CRC) is one of the most common malignancies and most of the patients diagnosed with advanced CRC have unsatisfactory treatment effect and poor prognosis. The purpose of this study was to investigate the effect of CCNI2 on the development of CRC. In this sutdy, immunohistochemical staining was used to detect CCNI2 expression levels in clinical samples, meanwhile, the Kaplan-Meier survival analysis was conducted. Celigo cell counting assay was used for screening shCCNI2s. QPCR and WB were performed to verify knockdown efficiency of CCNI2. Cell proliferation, colony formation, cell cycle, apoptosis, and mechanism investigation of CCNI2 knockdown were investigated by MTT assay, colony formation assay, fluorescence-activated cell sorting, and human apoptosis antibody array, respectively. Otherwise, the mouse model of CCNI2 knockdown was also constructed. The results of immunohistochemical staining and qPCR indicated that CCNI2 had a high expression level in the CRC tissues and cell lines. Kaplan-Meier survival analysis manifested that the high expression of CCNI2 suggested poor prognosis. The expression of CCNI2 was significantly reduced by CCNI2-siRNAs, and the downregulated expression level of CCNI2 inhibited CRC cell proliferation and colony formation, arrested cell cycle in G2 phase, as well as promoted cell apoptosis. The various indexes of solid tumor in mice models indicated that CCNI2 knockdown could suppress the growth of CRC tumor. Based on the comprehensive analysis of the above results, CCNI2 was contributed to the progression of CRC and could serve as a prognostic marker for CRC.


Subject(s)
Colorectal Neoplasms/metabolism , Cyclin I/metabolism , Aged , Animals , Apoptosis/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Cyclin I/genetics , Disease Models, Animal , Disease Progression , Down-Regulation , Female , G2 Phase , Gene Knockdown Techniques , Humans , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Polymerase Chain Reaction/methods , Tumor Stem Cell Assay
13.
Liver Int ; 30(1): 119-25, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19845855

ABSTRACT

BACKGROUND: The mammalian cyclin kinase subunit (Cks) family has two members, Cks1 and Cks2, which were identified based on the protein sequence homology to yeast Cks. Overexpression of Cks1 and Cks2 has been reported to be associated with high aggressiveness and a poor prognosis in various malignancies, including gastric, breast and prostate carcinomas. Yet, whether Cks1 and Cks2 are overexpressed in hepatocellular carcinoma (HCC) remains uncharacterized. AIMS: To investigate whether overexpression of the Cks family is clinically relevant to HCC, and whether expression patterns of Cks1 and Cks2 in HCC have diagnostic and prognostic value. METHODS: Real-time quantitative reverse transcriptase polymerase chain reaction, immunostaining and Western blot analyses were used to detect the expression of Cks1 and Cks2 at the mRNA and protein levels respectively. The associations between Cks1 and Cks2 expressions and clinical features, as well as the association between Cks1 or Cks2 and p27(kip1) expressions in HCC, were analysed. RESULTS: Expressions of Cks1 and Cks2 at both mRNA and protein levels were significantly higher in HCC than those in the adjacent noncancerous tissues (including chronic hepatitis and cirrhosis) and normal liver tissues. Overexpressions of Cks1 and Cks2 in HCC were closely associated with poor differentiation features. The expressions of both Cks1 and Cks2 were negatively associated with p27(kip1) at the protein level. CONCLUSIONS: Overexpression of Cks1 and Cks2 is associated with the aggressive tumour behaviours of HCC, and thus has diagnostic and prognostic value. Further efforts are needed to develop novel biomarkers for HCC based on CKs1 and Cks2 expressions.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Cyclin I/metabolism , Liver Neoplasms/enzymology , Protein Kinases/metabolism , Biomarkers, Tumor/metabolism , CDC2-CDC28 Kinases , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/genetics , Carrier Proteins/genetics , Cell Count , Cell Cycle Proteins/genetics , Cyclin I/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Hepatitis, Chronic/diagnosis , Hepatitis, Chronic/enzymology , Hepatitis, Chronic/genetics , Humans , Liver Cirrhosis/diagnosis , Liver Cirrhosis/enzymology , Liver Cirrhosis/genetics , Liver Neoplasms/diagnosis , Liver Neoplasms/genetics , Prognosis , Protein Kinases/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
14.
Medicine (Baltimore) ; 99(8): e19097, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32080083

ABSTRACT

Liquid biopsy is an emerging technique for noninvasive detection of various cancers. Majority of liquid biopsy tests still, however, use solitary type of biomarkers with unsatisfactory sensitivity and specificity. To this end, a combined approach of circulating tumor cells (CTCs) and salivary mRNA biomarkers was evaluated for discriminating non-small-cell lung cancer (NSCLC) from healthy controls.Our study included a discovery phase to find multiple biomarkers, and an independent validation phase to confirm the applicability of the selected biomarkers. In the discovery phase, CTC level in blood and 5 mRNA biomarkers in saliva (i.e., CCNI, Epidermal growth factor receptor [EGFR], FGF19, FRS2, and GREB1) were measured for 140 NSCLC patients and 140 healthy controls, followed by developing a predictive model. Next, this panel of biomarkers was applied to another patient cohort consisted of 60 patients with NSCLC and 60 healthy controls in the validation phase.We found that our novel biomarker panel could differentiate patients with NSCLC from healthy controls with high sensitivity (92.1%) and high specificity (92.9%) in the discovery phase. In the validation phase, we achieved sensitivity of 88.3% and specificity of 90.0%.To our best knowledge, it is the first time that a combined use of CTC and salivary mRNA biomarkers were applied for noninvasive detection of NSCLC.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Non-Small-Cell Lung/diagnosis , Neoplastic Cells, Circulating/metabolism , RNA, Messenger/metabolism , Saliva/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adult , Aged , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cyclin I/metabolism , ErbB Receptors/metabolism , Female , Fibroblast Growth Factors/metabolism , Humans , Liquid Biopsy/methods , Lung Neoplasms/diagnosis , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Membrane Proteins/metabolism , Middle Aged , Neoplasm Proteins/metabolism , Neoplastic Cells, Circulating/pathology , Predictive Value of Tests , Sensitivity and Specificity
15.
J Neurosci ; 27(34): 9201-19, 2007 Aug 22.
Article in English | MEDLINE | ID: mdl-17715356

ABSTRACT

The cellular pathways of motor neuronal injury have been investigated in the SOD1 G93A murine model of familial amyotrophic lateral sclerosis (ALS) using laser-capture microdissection and microarray analysis. The advantages of this study include the following: analysis of changes specifically in motor neurons (MNs), while still detecting effects of interactions with neighboring cells; the ability to profile changes during disease progression, an approach not possible in human ALS; and the use of transgenic mice bred on a homogeneous genetic background, eliminating the confounding effects arising from a mixed genetic background. By using this rigorous approach, novel changes in key cellular pathways have been detected at both the presymptomatic and late stages, which have been validated by quantitative reverse transcription-PCR. At the presymptomatic stage (60 d), MNs extracted from SOD1 G93A mice show a significant increase in expression of genes subserving both transcriptional and translational functions, as well as lipid and carbohydrate metabolism, mitochondrial preprotein translocation, and respiratory chain function, suggesting activation of a strong cellular adaptive response. Mice 90 d old still show upregulation of genes involved in carbohydrate metabolism, whereas transcription and mRNA processing genes begin to show downregulation. Late in the disease course (120 d), important findings include the following: marked transcriptional repression, with downregulation of multiple transcripts involved in transcriptional and metabolic functions; upregulation of complement system components; and increased expression of key cyclins involved in cell-cycle regulation. The changes described in the motor neuron transcriptome evolving during the disease course highlight potential novel targets for neuroprotective therapeutic intervention.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Gene Expression Regulation/genetics , Motor Neurons/metabolism , Superoxide Dismutase/genetics , Age Factors , Amyotrophic Lateral Sclerosis/genetics , Animals , Cyclin I , Cyclins/genetics , Cyclins/metabolism , Disease Models, Animal , Disease Progression , Malate Dehydrogenase/genetics , Malate Dehydrogenase/metabolism , Mice , Mice, Transgenic , Microarray Analysis/methods , Motor Neurons/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods
16.
Biochim Biophys Acta ; 1774(6): 764-71, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17507299

ABSTRACT

To identify new biomarkers that improve the early diagnosis and lead to possible therapeutic targets in pancreatic carcinoma, we performed a proteomic approach to compare serum protein expression patterns of pancreatic carcinoma patients with that of gastric cancer patients, other pancreatic disease patients, and healthy volunteers. By two-dimensional gel electrophoresis (2-DE) analyses and mass spectroscopic identification, 10 protein spots were found significantly changed in pancreatic carcinoma and 5 proteins including cyclin I, Rab GDP dissociation inhibitor beta (GDI2), alpha-1 antitrypsin precursor, Haptoglobin precursor, and Serotransferrin precursor were successfully identified. The increased levels of cyclin I and GDI2 found to be associated with pancreatic carcinoma were further confirmed by Western blot analyses in an independent series of serum samples and/or pancreatic juice samples. Applying immunohistochemistry, we further validated expression of cyclin I and GDI2 in additional pancreatic carcinomas. These results indicate that cyclin I and GDI2 may be potential molecular targets for pancreatic cancer diagnostics and therapeutics.


Subject(s)
Biomarkers, Tumor/blood , Blood Proteins/analysis , Cyclins/blood , Guanine Nucleotide Dissociation Inhibitors/blood , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/diagnosis , Biomarkers, Tumor/chemistry , Blood Proteins/chemistry , Blotting, Western , Cyclin I , Cyclins/chemistry , Electrophoresis, Gel, Two-Dimensional , Guanine Nucleotide Dissociation Inhibitors/chemistry , Health , Humans , Immunohistochemistry , Middle Aged , Proteomics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
17.
Nat Commun ; 9(1): 3919, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30254248

ABSTRACT

In addition to genomic mutations, RNA editing is another major mechanism creating sequence variations in proteins by introducing nucleotide changes in mRNA sequences. Deregulated RNA editing contributes to different types of human diseases, including cancers. Here we report that peptides generated as a consequence of RNA editing are indeed naturally presented by human leukocyte antigen (HLA) molecules. We provide evidence that effector CD8+ T cells specific for edited peptides derived from cyclin I are present in human tumours and attack tumour cells that are presenting these epitopes. We show that subpopulations of cancer patients have increased peptide levels and that levels of edited RNA correlate with peptide copy numbers. These findings demonstrate that RNA editing extends the classes of HLA presented self-antigens and that these antigens can be recognised by the immune system.


Subject(s)
Antigens, Neoplasm/immunology , Epitopes/immunology , Immune System/immunology , Neoplasms/immunology , RNA Editing/immunology , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cells, Cultured , Cyclin I/genetics , Cyclin I/immunology , Cyclin I/metabolism , Cytotoxicity, Immunologic/immunology , HLA Antigens/immunology , Humans , Neoplasms/genetics , Neoplasms/metabolism , Peptides/genetics , Peptides/immunology , Peptides/metabolism , Proteogenomics/methods
18.
Sci Rep ; 7: 40979, 2017 01 23.
Article in English | MEDLINE | ID: mdl-28112194

ABSTRACT

In contrast to conventional cyclin-dependent kinases that are important for mitotic cell division, cyclin-dependent kinase 5 (CDK5) is predominantly activated in post-mitotic cells and is involved in various cellular events. The kinase activity of CDK5 is tightly regulated by specific activators including p35, p39, and cyclin I (CCNI). Here we show that cyclin I-like (CCNI2), a homolog of CCNI, interacts with CDK5 and activates the kinase activity of CDK5. Different from CCNI, which colocalizes with CDK5 in the nuclei in transfected cells, CCNI2 mainly retains CDK5 in the cytoplasm as well as on the cell membrane. Furthermore, although the expression level of CCNI2 mRNA and CCNI2 protein do not change significantly during cell cycle, depletion of CCNI2 with siRNA affects cell cycle progression as well as cell proliferation. In conclusion, our data strongly suggest that CCNI2 is a novel CDK5 activator and is involved in cell cycle regulation.


Subject(s)
Cell Cycle , Cyclin I/metabolism , Cyclin-Dependent Kinase 5/metabolism , Gene Expression Regulation , A549 Cells , Blotting, Western , Cell Proliferation , Gene Expression Profiling , HeLa Cells , Humans , Protein Interaction Mapping , Reverse Transcriptase Polymerase Chain Reaction , Two-Hybrid System Techniques
19.
Anticancer Res ; 35(2): 1115-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25667501

ABSTRACT

BACKGROUND/AIM: Ovarian cancer is the second most common gynecological malignancy after cancer of the uterine corpus, and the fifth leading cause of cancer-related death among women. It has been discovered that cyclin I (CCNI) protein expression correlates with the proliferation of cancer cells and expression of angiogenesis-related proteins, such as vascular endothelial growth factor (VEGF) and VEGF receptor 2/kinase insert domain receptor (VEGFR2/KDR). We examined whether any association exists between mRNA expression of CCNI and KDR genes in epithelial ovarian cancer (EOC) tissues, clinicopathological parameters and patients' response to chemotherapy. MATERIALS AND METHODS: Expression of CCNI and KDR genes was analyzed by quantitative real-time reverse transcription PCR in 40 human primary EOC tissues and four human ovarian cancer cell lines (TOV-112D, OV-90, OVCAR-3 and Caov-3). RESULTS: CCNI and KDR mRNA expression was detected in all EOC tissues and ovarian cancer cell lines. The mRNA levels of both genes were significantly higher in EOC than in ovarian cancer cell lines (p<0.001). Neither CCNI nor KDR mRNA expression in EOC tissues was significantly associated with variables such as age, menopausal status, International Federation of Gynecology and Obstetrics (FIGO) stage, residual disease, patients' response to chemotherapy, tumor histology, grade or sensitivity to chemotherapy. However, we demonstrated a significant positive correlation between the mRNA expression of KDR and CCNI in EOC tissues (R=0.530, p<0.001). CONCLUSION: Neither CCNI nor KDR mRNA expression predicts response of patients with EOC to platinum-based first-line chemotherapy. Cyclin I may be involved in angiogenesis in EOC, which needs further investigation.


Subject(s)
Cyclin I/genetics , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , RNA, Messenger/genetics , Base Sequence , Carcinoma, Ovarian Epithelial , DNA Primers , Female , Humans , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/metabolism
20.
Eur Rev Med Pharmacol Sci ; 19(23): 4533-41, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26698249

ABSTRACT

OBJECTIVE: Cisplatin (cis-diamminedichloroplatinum II, CDDP) is one of the most effective chemotherapeutic agents and is widely used in the treatment of cervical cancer (CC), but cancer cell acquired resistance to this drug during the course of its treatment. The aim of this study was to investigate the role of cyclin I to cisplatin resistance in CC cell. PATIENTS AND METHODS: Cervical tumor specimens from 30 patients were recruited in this study. We analyzed the expression of cyclin I by real-time polymerase chain reaction (qRT-PCR), Western blotting examination of downstream effectors. Cell proliferation assay and xenograft experiments were performed for cisplatin cytotoxicity assay. Lentivirus-mediated and siRNA-mediated genes overexpression or knockdown were applied to investigate the role of cyclin I to cisplatin resistance in CC cell. RESULTS: We found that high level of cyclin I was associated with cisplatin resistance in CC. Here, we described that cyclin I protein becomes highly expressed in human CC patients resistant to cisplatin chemotherapy. Stable overexpressed cyclin I promotes Hela cell resistance to higher concentrations of cisplatin. In addition, upregulated level of cyclin I increased tumor cells growth in vitro and enhanced tumor resistance to cisplatin in vivo. The further mechanism investigated showed that cyclin I upregulated the expression of cyclin-dependent kinase 5 (Cdk5) promoting cisplatin resistance by preventing apoptosis in CC cell line. Consistently, the cyclin I overexpressed Hela cell lines produce increased sensitivity to cisplatin treatment through knockdown of Cdk5 protein with siRNA. CONCLUSIONS: These data suggest that a cyclin I-Cdk5 complex forms a critical antiapoptotic factor in the process of generating cisplatin resistance in cervical cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/genetics , Cisplatin/therapeutic use , Cyclin I/genetics , Cyclin-Dependent Kinase 5/genetics , Drug Resistance, Neoplasm/genetics , Uterine Cervical Neoplasms/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Squamous Cell/drug therapy , Cyclin I/antagonists & inhibitors , Cyclin-Dependent Kinase 5/metabolism , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Small Interfering/pharmacology , Uterine Cervical Neoplasms/drug therapy , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL