Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 304
1.
Endocrinology ; 163(3)2022 03 01.
Article En | MEDLINE | ID: mdl-35038735

CONTEXT: 3,5,3'-L-triiodothyronine (T3) is a potent inducer of hepatocyte proliferation via the Wnt/ß-catenin signaling pathway. Previous studies suggested the involvement of rapid noncanonical thyroid hormone receptor (TR) ß signaling, directly activating hepatic Wnt/ß-catenin signaling independent from TRß DNA binding. However, the mechanism by which T3 increases Wnt/ß-catenin signaling in hepatocytes has not yet been determined. OBJECTIVE: We aimed to determine whether DNA binding of TRß is required for stimulation of hepatocyte proliferation by T3. METHODS: Wild-type (WT) mice, TRß knockout mice (TRß KO), and TRß mutant mice with either specifically abrogated DNA binding (TRß GS) or abrogated direct phosphatidylinositol 3 kinase activation (TRß 147F) were treated with T3 for 6 hours or 7 days. Hepatocyte proliferation was assessed by Kiel-67 (Ki67) staining and apoptosis by terminal deoxynucleotidyl transferase dUTP nick-end labeling assay. Activation of ß-catenin signaling was measured in primary murine hepatocytes. Gene expression was analyzed by microarray, gene set enrichment analysis (GSEA), and quantitative reverse transcription polymerase chain reaction. RESULTS: T3 induced hepatocyte proliferation with an increased number of Ki67-positive cells in WT and TRß 147F mice (9.2% ±â€…6.5% and 10.1% ±â€…2.9%, respectively) compared to TRß KO and TRß GS mice (1.2% ±â€…1.1% and 1.5% ±â€…0.9%, respectively). Microarray analysis and GSEA showed that genes of the Wnt/ß-catenin pathway-among them, Fzd8 (frizzled receptor 8) and Ctnnb1 (ß-catenin)-were positively enriched only in T3-treated WT and TRß 147F mice while B-cell translocation gene anti-proliferation factor 2 was repressed. Consequently, expression of Ccnd1 (CyclinD1) was induced. CONCLUSIONS: Instead of directly activating Wnt signaling, T3 and TRß induce key genes of the Wnt/ß-catenin pathway, ultimately stimulating hepatocyte proliferation via CyclinD1. Thus, canonical transcriptional TRß action is necessary for T3-mediated stimulation of hepatocyte proliferation.


Cell Proliferation/physiology , Hepatocytes/physiology , Thyroid Hormone Receptors beta/physiology , Triiodothyronine/pharmacology , Animals , Binding Sites/genetics , Cell Proliferation/drug effects , Cyclin D1/physiology , DNA/metabolism , Gene Expression/drug effects , Hepatocytes/drug effects , Hypothyroidism , Male , Mice , Mice, Knockout , Mice, Mutant Strains , Mutation , Signal Transduction/drug effects , Signal Transduction/physiology , Thyroid Hormone Receptors beta/genetics , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics
2.
J BUON ; 26(3): 882-888, 2021.
Article En | MEDLINE | ID: mdl-34268949

PURPOSE: This study aims to elucidate the biological functions of CDCA2 (cell division cycle associated 2) in hepatocellular carcinoma (HCC) progression and the potential mechanism. METHODS: CDCA2 levels in HCC tissues and cell lines were detected by quantitative real-time polymerase chain reaction (qRT-PCR). The relationship between CDCA2 and clinical characteristics in HCC patients was analyzed. Cox proportional-hazards model was applied for assessing the potential factors influencing overall survival in HCC. Three CDCA2 siRNAs were generated and the most effective one was used in the following experiments. After knockdown of CDCA2 in HCC-LM3 cells, clonality and viability were examined. Meanwhile, cell cycle progression was detected by flow cytometry. Relative levels of CDCA2, p21, p27, CDK2, CCND1, CCNE1 and CCNB1 in HCC-LM3 cells were determined by qRT-PCR. The activation of the protein kinase B (Akt) signaling was examined by Western blot. Subsequently, we constructed HCC xenograft model in nude mice. Tumor volume and tumor weight of xenografted HCC were recorded. RESULTS: CDCA2 was upregulated in HCC tissues than that of para-tumor ones, especially HCC tissues with larger than 5 cm in tumor size or vascular invasion. CDCA2 level was related to tumor size, vascular invasion and tumor differentiation in HCC. Knockdown of CDCA2 inhibited clonality and viability in HCC-LM3 cells, and arrested cell cycle progression in G1 phase via downregulating CCND1. The phosphatidilinositol 3-kinase (PI3K)/Akt was activated by CDCA2 during the progression of HCC. Tumor volume and tumor weight of xenografted HCC decreased in nude mice with in vivo knockdown of CDCA2. CONCLUSIONS: CDCA2 triggers proliferative potential in HCC by targeting CCND1 via activating the PI3K/Akt signaling.


Carcinoma, Hepatocellular/pathology , Carrier Proteins/physiology , Cell Cycle Proteins/physiology , Cell Proliferation , Cyclin D1/physiology , Liver Neoplasms/pathology , Nuclear Proteins/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction , Animals , Disease Progression , Female , Humans , Male , Mice , Middle Aged , Tumor Cells, Cultured
3.
Andrology ; 9(6): 1923-1933, 2021 11.
Article En | MEDLINE | ID: mdl-34185441

BACKGROUND: The tricho-rhino-phalangeal syndrome-1 gene (Trps1) is an atypical GATA family member. Although current studies of Trps1 mainly focus on tumors, whether Trps1 plays a role in the male reproductive system remains unknown. OBJECTIVES: The purpose of this study was to elucidate the function of Trps1 in Leydig cells, indicating its regulatory mechanism on the cell cycle. METHODS: Gene-silencing technology, RNA-seq, RT-qPCR, and western blotting were used to evaluate the function of Trps1 in mouse primary Leydig cells and MLTC-1 cells. In addition, ChIP-base sets and ChIP-qPCR were employed to further assess the regulatory mechanism of Trps1 in MLTC-1 cells. RESULTS: Knockdown of Trps1 in Leydig cells significantly suppressed phosphorylation of Src and Akt and expression of Ccnd1, which was accompanied by impairment of cell proliferative ability. Trps1 may affect the cell cycle through the Src/Akt/Ccnd1 signaling pathway. In addition, Trps1 may bind to the promoter of Srcin1 to regulate its transcription, thus influencing Src phosphorylation levels and the proliferation of Leydig cells. DISCUSSION AND CONCLUSION: Src increases in Leydig cells during pubertal development, suggesting its functional involvement in differentiated adult Leydig cells. Inhibition of the Src/Akt pathway would reduce Ccnd1 expression. In the present study, we found that Trps1 may regulate the phosphorylation level of Src and Akt through Srcin1, targeting Ccnd1 to influence mouse Leydig cell proliferation. These findings shed light on the regulation of Trps1 on cell proliferation and differentiation of mouse Leydig cells.


Cell Proliferation/genetics , Cyclin D1/physiology , Leydig Cells/metabolism , Repressor Proteins/physiology , Animals , Cell Cycle/genetics , Cell Differentiation/genetics , Male , Mice , Signal Transduction/genetics
4.
Anticancer Res ; 41(3): 1299-1305, 2021 Mar.
Article En | MEDLINE | ID: mdl-33788721

BACKGROUND/AIM: A xanthophyll of fucoxanthin (Fx) is a potential chemopreventive agent. Familial adenomatous polyposis (FAP) is an inherited disease that is associated with a high risk of developing colorectal cancer. However, it remains unclear whether Fx can modify colorectal tumorigenesis in ApcMin/+ mice, a model mouse for human FAP. MATERIALS AND METHODS: We investigated the chemopreventive effect of Fx in dextran sodium sulfate (DSS)-treated ApcMin/+ mice. RESULTS: Administration of Fx in the diet for 5 weeks significantly suppressed the number of colorectal adenocarcinomas in DSS-treated male ApcMin/+ mice, although the treatment did not affect the occurrence of colorectal dysplastic crypts and adenoma in the mice. In addition, Fx down-regulated cyclin D1 expression (0.6-fold) in colorectal mucosa of ApcMin/+ mice when compared with that of the control mice. CONCLUSION: Fx possesses chemopreventive potential against progression of colorectal carcinogenesis in ApcMin/+ mice that receive inflammatory stimuli.


Adenomatous Polyposis Coli/complications , Anticarcinogenic Agents/administration & dosage , Colorectal Neoplasms/prevention & control , Xanthophylls/administration & dosage , Animals , Colorectal Neoplasms/chemically induced , Cyclin D1/analysis , Cyclin D1/physiology , Dextran Sulfate , Disease Models, Animal , Male , Mice
5.
Clin Nutr ; 40(4): 2025-2034, 2021 04.
Article En | MEDLINE | ID: mdl-33008652

BACKGROUND & AIMS: Our previous study found that platelet counts were positively associated with body fat percentage in human. In the present study, we conducted a reverse translational study to explore the role of platelets in modulating pre-adipocyte proliferation in mice. METHODS: Mouse pre-adipocyte cell line (3T3-L1) and human pre-adipocytes harvested from female subcutaneous fat were used. Pre-adipocytes were co-cultured with platelets or platelet releasate, which were isolated from mice or humans. The cell viability and proliferative ability of the pre-adipocytes were examined by MTT and flow cytometry assays. Western blotting analysis was used to determine the phosphorylation levels of proteins in the mTOR pathway. RESULTS: The number of platelets in the adipose tissues from obese mice was significantly higher than that from lean mice. Platelets and collagen-activated platelet releasate stimulated the proliferation of human pre-adipocytes and 3T3-L1 cells in vitro. Besides, platelets from obese mice were more potent in stimulating pre-adipocyte proliferation than those from lean control mice. Mechanistically, platelets enhanced pre-adipocyte proliferation through the acceleration of cell cycle progression from G0/G1 to S phase cell cycle progression. At the molecular level, platelets promoted pre-adipocyte proliferation through mTOR pathway-mediated upregulation of cyclin D1 expression. CONCLUSION: In conclusion, platelets and platelet releasate play an important role in the proliferation of pre-adipocytes. Our study may provide new clues and the molecular mechanism of the causal pathways between platelets and body fat to explain the finding we observed in population study.


Adipocytes/physiology , Adipose Tissue/physiopathology , Blood Platelets/physiology , 3T3-L1 Cells , Adipose Tissue/pathology , Animals , Apoptosis , Blood Platelets/pathology , Cell Communication , Cell Proliferation , Cell Survival , Cells, Cultured , Cyclin D1/physiology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Obesity/pathology , Obesity/physiopathology , Platelet Count , Specific Pathogen-Free Organisms , Subcutaneous Fat , TOR Serine-Threonine Kinases/physiology , Translational Research, Biomedical
6.
Rejuvenation Res ; 23(3): 262-265, 2020 Jun.
Article En | MEDLINE | ID: mdl-32484032

Exercise has long been known to extend health and lifespan in humans and other mammals. However, typically exercise is thought to slow the loss of function that accompanies aging. Brett et al. have now shown that exercise restores functional competency to regenerate muscle stem cells (MuSCs) in mice as well as restore a significant portion of the transcriptional signature associated with young MuSCs. The mechanism involves the likely induction of plasma-borne factors that upregulate cell cycle regulator cyclin D1, which otherwise decreases with increasing age. Cyclin D1, in turn, through its noncanonical attenuation of TGF-beta/Smad3 signaling, helps maintain the regenerative capacity of MuSCs, which is lost as TGF-beta signaling increases with age. Interestingly, elevated levels of some proinflammatory regulators including NF-κB, TNF-alpha, and interleukin 6 (IL-6) are also reduced by exercise or ectopic expression of cyclin D1. Importantly, the rejuvenation is not complete, as Notch signaling, which also decreases with age, remains at old levels and the rejuvenative effect is not permanent: wearing off in ∼2 weeks after cessation of exercise. Understanding the limitations of the rejuvenative effect of exercise on MuSCs at the molecular level, including changes in the epigenome such as altered DNA methylation age, will be critical in developing more significant rejuvenative therapies including some for aged people wherein morbidities limit exercise.


Exercise/physiology , Muscle, Skeletal/physiology , Physical Conditioning, Animal/physiology , Rejuvenation/physiology , Stem Cells/physiology , Aging/physiology , Animals , Cyclin D1/physiology , Frailty/physiopathology , Frailty/therapy , Humans , Mice , Muscle, Skeletal/cytology , NF-kappa B/metabolism , Regeneration/physiology , Signal Transduction/physiology , Stem Cells/cytology , Tumor Necrosis Factor-alpha/metabolism
7.
Nat Commun ; 11(1): 2898, 2020 06 09.
Article En | MEDLINE | ID: mdl-32518258

The sequential generation of layer-specific cortical neurons requires radial glia cells (RGCs) to precisely balance self-renewal and lineage commitment. While specific cell-cycle phases have been associated with these decisions, the mechanisms linking the cell-cycle machinery to cell-fate commitment remain obscure. Using single-cell RNA-sequencing, we find that the strongest transcriptional signature defining multipotent RGCs is that of G2/M-phase, and particularly CYCLIN-B1/2, while lineage-committed progenitors are enriched in G1/S-phase genes, including CYCLIN-D1. These data also reveal cell-surface markers that allow us to isolate RGCs and lineage-committed progenitors, and functionally confirm the relationship between cell-cycle phase enrichment and cell fate competence. Finally, we use cortical electroporation to demonstrate that CYCLIN-B1/2 cooperate with CDK1 to maintain uncommitted RGCs by activating the NOTCH pathway, and that CYCLIN-D1 promotes differentiation. Thus, this work establishes that cell-cycle phase-specific regulators act in opposition to coordinate the self-renewal and lineage commitment of RGCs via core stem cell regulatory pathways.


Cyclin B1/physiology , Cyclin B2/physiology , Cyclin D1/physiology , Gene Expression Regulation, Developmental , Animals , CDC2 Protein Kinase/physiology , Cell Cycle , Cell Differentiation , Cell Lineage , Cell Separation , Cerebral Cortex/embryology , Female , Flow Cytometry , Mice , Mice, Inbred C57BL , Neuroglia/metabolism , Sequence Analysis, RNA , Signal Transduction , Stem Cells/cytology
8.
J Pharmacol Sci ; 143(3): 219-225, 2020 Jul.
Article En | MEDLINE | ID: mdl-32418739

OBJECTIVE: Chemoresistance is one of the major barriers in chemotherapy-based hepatocellular carcinoma (HCC) intervention. 5-Fluorouracil (5-Fu) is a widely used as an anticancer drug. Liver cancer stem cells (LCSCs) are considered the origin of tumor recurrence and resistance. CCND1 (Cyclin D1) plays an important role in tumorigenesis and metastasis in multiple cancers including HCC. Herein, this study was designed to explore the role of CCND1 in regulating LCSCs differentiation and 5-Fu resistance in HCC cells. METHODS: The CCND1 mRNA level was examined by qRT-PCR. The protein levels of γ-H2AX (a DNA damage marker) and RAD51 (a DNA repair protein) were examined by Western blot. CD133 was used as a LCSC marker and CD133+ cell percentage in HCC cells was detected by flow cytometry. RESULTS: CCND1 silencing decreased CD133+ cell percentage in HepG2 and SMMC-7721 cells. Furthermore, CCND1 silencing significantly increased protein level of γ-H2AX and decreased that of RAD51 under 5-Fu exposure. Moreover, CCND1 silencing enhanced the sensitivity of HepG2 and SMMC-7721 cells to 5-Fu, which was effectively abrogated by RAD51 upregulation. CONCLUSION: Collectively, CCND1 silencing suppresses LCSCs differentiation and overcomes 5-Fu resistance in HCC.


Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Differentiation/genetics , Cyclin D1/genetics , Cyclin D1/physiology , Drug Resistance, Neoplasm/genetics , Fluorouracil/pharmacology , Gene Silencing , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Neoplastic Stem Cells/pathology , Cell Culture Techniques , Hep G2 Cells , Humans , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Up-Regulation/genetics
9.
Oncogene ; 38(39): 6615-6629, 2019 09.
Article En | MEDLINE | ID: mdl-31371779

Oncogenic KIT or PDGFRA tyrosine kinase mutations are compelling therapeutic targets in most gastrointestinal stromal tumors (GISTs), and the KIT inhibitor, imatinib, is therefore standard of care for patients with metastatic GIST. However, some GISTs lose expression of KIT oncoproteins, and therefore become KIT-independent and are consequently resistant to KIT-inhibitor drugs. We identified distinctive biologic features in KIT-independent, imatinib-resistant GISTs as a step towards identifying drug targets in these poorly understood tumors. We developed isogenic GIST lines in which the parental forms were KIT oncoprotein-dependent, whereas sublines had loss of KIT oncoprotein expression, accompanied by markedly downregulated expression of the GIST biomarker, protein kinase C-theta (PRKCQ). Biologic mechanisms unique to KIT-independent GISTs were identified by transcriptome sequencing, qRT-PCR, immunoblotting, protein interaction studies, knockdown and expression assays, and dual-luciferase assays. Transcriptome sequencing showed that cyclin D1 expression was extremely low in two of three parental KIT-dependent GIST lines, whereas cyclin D1 expression was high in each of the KIT-independent GIST sublines. Cyclin D1 inhibition in KIT-independent GISTs had anti-proliferative and pro-apoptotic effects, associated with Rb activation and p27 upregulation. PRKCQ, but not KIT, was a negative regulator of cyclin D1 expression, whereas JUN and Hippo pathway effectors YAP and TAZ were positive regulators of cyclin D1 expression. PRKCQ, JUN, and the Hippo pathway coordinately regulate GIST cyclin D1 expression. These findings highlight the roles of PRKCQ, JUN, Hippo, and cyclin D1 as oncogenic mediators in GISTs that have converted, during TKI-therapy, to a KIT-independent state. Inhibitors of these pathways could be effective therapeutically for these now untreatable tumors.


Cyclin D1/physiology , Gastrointestinal Stromal Tumors/genetics , Proto-Oncogene Proteins c-kit/genetics , Antineoplastic Agents/therapeutic use , Cell Proliferation/physiology , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/metabolism , Humans , Protein Kinase C-theta/physiology , Protein Kinase Inhibitors/therapeutic use
10.
EMBO J ; 38(6)2019 03 15.
Article En | MEDLINE | ID: mdl-30643018

Adult neurogenesis is involved in cognitive performance but studies that manipulated this process to improve brain function are scarce. Here, we characterized a genetic mouse model in which neural stem cells (NSC) of the subventricular zone (SVZ) were temporarily expanded by conditional expression of the cell cycle regulators Cdk4/cyclinD1, thus increasing neurogenesis. We found that supernumerary neurons matured and integrated in the olfactory bulb similarly to physiologically generated newborn neurons displaying a correct expression of molecular markers, morphology and electrophysiological activity. Olfactory performance upon increased neurogenesis was unchanged when mice were tested on relatively easy tasks using distinct odor stimuli. In contrast, intriguingly, increasing neurogenesis improved the discrimination ability of mice when challenged with a difficult task using mixtures of highly similar odorants. Together, our study provides a mammalian model to control the expansion of somatic stem cells that can in principle be applied to any tissue for basic research and models of therapy. By applying this to NSC of the SVZ, we highlighted the importance of adult neurogenesis to specifically improve performance in a challenging olfactory task.


Discrimination Learning , Neural Stem Cells/physiology , Neurogenesis/physiology , Odorants/analysis , Olfactory Bulb/physiology , Animals , Cyclin D1/physiology , Cyclin-Dependent Kinase 4/physiology , Disease Models, Animal , Male , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Olfactory Bulb/cytology , Olfactory Bulb/drug effects
11.
Gynecol Oncol ; 152(1): 157-165, 2019 01.
Article En | MEDLINE | ID: mdl-30414739

OBJECTIVE: Poly(ADP-ribose) polymerase inhibitors (PARPi) are active in cancer cells that have impaired repair of DNA by the homologous recombination (HR) pathway. Strategies that disrupt HR may sensitize HR-proficient tumors to PARP inhibition. As a component of the core cell cycle machinery, cyclin D1 has unexpected function in DNA repair, suggesting that targeting cyclin D1 may represent a plausible strategy for expanding the utility of PARPi in ovarian cancer. METHODS: BRCA1 wildtype ovarian cancer cells (A2780 and SKOV3) were treated with a combination of CCND1 siRNA and olaparib in vitro. Cell viability was assessed by MTT. The effects of the combined treatment on DNA damage repair and cell cycle progression were examined to dissect molecular mechanisms. In vivo studies were performed in an orthotopic ovarian cancer mouse model. Animals were treated with a combination of lentivirus-mediated CCND1 shRNA and olaparib or olaparib plus scrambled shRNA. Molecular downstream effects were examined by immunohistochemistry. RESULTS: Silencing of cyclin D1 sensitized ovarian cancer cells to olaparib through interfering with RAD51 accumulation and inducing cell cycle G0/G1 arrest. Treatment of lentivirus-mediated CCND1-shRNA in nude mice statistically significantly augmented the olaparib response (mean tumor weight ±â€¯SD, CCND1-shRNA plus olaparib vs scrambled shRNA plus olaparib: 0.172 ±â€¯0.070 g vs 0.324 ±â€¯0.044 g, P< 0.05). CONCLUSIONS: Silencing of cyclin D1 combined with olaparib may lead to substantial benefit for ovarian cancer management by mimicking a BRCAness phenotype, and induction of G0/G1 cell cycle arrest.


Cyclin D1/physiology , DNA Breaks, Double-Stranded , DNA Repair , Genes, BRCA1 , Ovarian Neoplasms/drug therapy , Phthalazines/therapeutic use , Piperazines/therapeutic use , Animals , Cell Cycle , Cell Line, Tumor , Cyclin D1/genetics , Female , Humans , Mice , Mice, Inbred BALB C , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Rad51 Recombinase/analysis
12.
Arterioscler Thromb Vasc Biol ; 39(1): 48-62, 2019 01.
Article En | MEDLINE | ID: mdl-30580569

Objective- Hypoxic pulmonary hypertension (HPH) is characterized by proliferative vascular remodeling. Abnormal pulmonary artery smooth muscle cells proliferation and endothelial dysfunction are the primary cellular bases of vascular remodeling. AQP1 (aquaporin-1) is regulated by oxygen level and has been observed to play a role in the proliferation and migration of pulmonary artery smooth muscle cells. The role of AQP1 in HPH pathogenesis has not been directly determined to date. To determine the possible roles of AQP1 in the pathogenesis of HPH and explore its possible mechanisms. Approach and Results- Aqp1 knockout mice were used, and HPH model was established in this study. Primary pulmonary artery smooth muscle cells, primary mouse lung endothelial cells, and lung tissue sections from HPH model were used. Immunohistochemistry, immunofluorescence and Western blot, cell cycle, apoptosis, and migration analysis were performed in this study. AQP1 expression was upregulated by chronic hypoxia exposure, both in pulmonary artery endothelia and medial smooth muscle layer of mice. Aqp1 deficiency attenuated the elevation of right ventricular systolic pressures and mitigated pulmonary vascular structure remodeling. AQP1 deletion reduced abnormal cell proliferation in pulmonary artery and accompanied with accumulation of HIF (hypoxia-inducible factor). In vitro, Aqp1 deletion reduced hypoxia-induced proliferation, apoptosis resistance, and migration ability of primary cultured pulmonary artery smooth muscle cells and repressed HIF-1α protein stability. Furthermore, Aqp1 deficiency protected lung endothelial cells from apoptosis in response to hypoxic injury. Conclusions- Our data showed that Aqp1 deficiency could attenuate hypoxia-induced vascular remodeling in the development of HPH. AQP1 may be a potential target for pulmonary hypertension treatment.


Aquaporin 1/physiology , Hypertension, Pulmonary/etiology , Hypoxia/complications , Animals , Aquaporin 1/genetics , Cells, Cultured , Cyclin D1/physiology , Hypertension, Pulmonary/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Male , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/physiology , Vascular Remodeling
13.
Eur Rev Med Pharmacol Sci ; 22(17): 5588-5596, 2018 09.
Article En | MEDLINE | ID: mdl-30229833

OBJECTIVE: Chemokine (C-C motif) ligand 2 (CCL2) is a member of the CC subfamily, which displays chemotactic activity for monocytes and basophils. This molecule plays a very important role in many solid tumors and shows changes in the bone marrow microenvironment. However, its role in acute myeloid leukaemia (AML) is still unclear. MATERIALS AND METHODS: In this study, we established a HL-60 cell line with CCL2 knockdown to explore its effect on leukemogenesis. Lentivirus with CCL2-knockdown was successfully constructed after screening effective CCL2 short hairpin RNA (shRNA) sequences and was transfected into HL-60 cells, which was further validated at the mRNA and protein levels by real-time polymerase chain reaction (PCR) and Western blotting, respectively. RESULTS: Low expression of CCL2 significantly decreased HL-60 cell growth by increasing the cell arrest at G1 phase by 12% more than controls. We applied RNA sequencing technology to discriminate the gene expression profiles between the cells with CCL2 knockdown and the controls, and Cyclin D1 was selected for further experiments as its expression level was significantly downregulated, which was validated at the mRNA and protein levels. Cyclin D1 knockdown experiments showed that the cell proliferation rate was evidently decelerated, and cell cycle analysis also indicated a similar pattern for CCL2. CONCLUSIONS: Our study revealed that Cyclin D1 is an effector that mediates CCL2's function in cell proliferation by blocking cells at G1 phase.


Chemokine CCL2/physiology , Cyclin D1/physiology , Leukemia, Myeloid, Acute/pathology , Cell Line, Tumor , Cell Proliferation , HL-60 Cells , Humans
14.
PLoS One ; 12(2): e0172613, 2017.
Article En | MEDLINE | ID: mdl-28222172

OBJECTIVE: Epidermal stem cells (ESCs) play a critical role in wound healing, but the mechanism underlying ESC proliferation is not well defined. Here, we explore the effects of RhoA on ESC proliferation and the possible underlying mechanism. METHODS: Human ESCs were enriched by rapid adhesion to collagen IV. RhoA(+/+)(G14V), RhoA(-/-)(T19N) and pGFP control plasmids were transfected into human ESCs. The effect of RhoA on cell proliferation was detected by cell proliferation and DNA synthesis assays. Induction of PKN1 activity by RhoA was determined by immunoblot analysis, and the effects of PKN1 on RhoA in terms of inducing cell proliferation and cyclin D1 expression were detected using specific siRNA targeting PKN1. The effects of U-46619 (a RhoA agonist) and C3 transferase (a RhoA antagonist) on ESC proliferation were observed in vivo. RESULTS: RhoA had a positive effect on ESC proliferation, and PKN1 activity was up-regulated by the active RhoA mutant (G14V) and suppressed by RhoA T19N. Moreover, the ability of RhoA to promote ESC proliferation and DNA synthesis was interrupted by PKN1 siRNA. Additionally, cyclin D1 protein and mRNA expression levels were up-regulated by RhoA G14V, and these effects were inhibited by siRNA-mediated knock-down of PKN1. RhoA also promoted ESC proliferation via PKN in vivo. CONCLUSION: This study shows that the effect of RhoA on ESC proliferation is mediated by activation of the PKN1-cyclin D1 pathway in vitro, suggesting that RhoA may serve as a new therapeutic target for wound healing.


Cyclin D1/physiology , Epidermal Cells , Epithelial Cells/metabolism , Protein Kinase C/physiology , Signal Transduction/physiology , Stem Cells/metabolism , Wound Healing , rhoA GTP-Binding Protein/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , ADP Ribose Transferases/pharmacology , Animals , Botulinum Toxins/pharmacology , Burns/physiopathology , Burns/therapy , Carbazoles/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Cyclin D1/biosynthesis , Cyclin D1/genetics , DNA Replication/drug effects , Epithelial Cells/cytology , Humans , Indole Alkaloids/pharmacology , Male , Mice , Mice, Inbred C57BL , Mutation, Missense , Primary Cell Culture , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , RNA Interference , RNA, Small Interfering , Random Allocation , Stem Cells/cytology , Transfection , Wound Healing/physiology , rho GTP-Binding Proteins/agonists , rho GTP-Binding Proteins/antagonists & inhibitors , rhoA GTP-Binding Protein/deficiency , rhoA GTP-Binding Protein/genetics
15.
Apoptosis ; 22(4): 570-584, 2017 Apr.
Article En | MEDLINE | ID: mdl-28176146

Centchroman (CC) or Ormeloxifene has been shown to induce apoptosis and cell cycle arrest in various types of cancer cells. This has, however, not been addressed for endometrial cancer cells where its (CC) mechanism of action remains unclear. This study focuses on the basis of antineoplasticity of CC by blocking the targets involved in the cell cycle, survival and apoptosis in endometrial cancer cells. Ishikawa Human Endometrial Cancer Cells were cultured under estrogen deprived medium, exposed to CC and analyzed for proliferation and apoptosis. Additionally, we also analyzed oxidative stress induced by CC. Cell viability studies confirmed the IC50 of CC in Ishikawa cells to be 20 µM after 48 h treatment. CC arrests the cells in G0/G1 phase through cyclin D1 and cyclin E mediated pathways. Phosphatidylserine externalization, nuclear morphology changes, DNA fragmentation, PARP cleavage, and alteration of Bcl-2 family protein expression clearly suggest ongoing apoptosis in the CC treated cells. Activation of caspase 3 & 9, up-regulation of AIF and inhibition of apoptosis by z-VAD-fmk clearly explains the participation of the intrinsic pathway of programmed cell death. Further, the increase of ROS, loss of MMP, inhibition of antioxidant (MnSOD, Cu/Zn-SOD and GST) and inhibition of apoptosis with L-NAC suggests CC induced oxidative stress leading to apoptosis via mitochondria mediated pathway. Therefore, CC could be a potential therapeutic agent for the treatment of Endometrial Cancer adjunct to its utility as a contraceptive and an anti-breast cancer agent.


Adenocarcinoma/pathology , Antineoplastic Agents, Hormonal/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Centchroman/pharmacology , Endometrial Neoplasms/pathology , Caspases/physiology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cyclin D1/physiology , Cyclin E/physiology , DNA Fragmentation/drug effects , Female , Humans , Inhibitory Concentration 50 , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Membrane Potential, Mitochondrial/drug effects , Neoplasm Proteins/physiology , Oxidation-Reduction , Protein Transport/drug effects
16.
Adv Clin Exp Med ; 26(9): 1335-1342, 2017 Dec.
Article En | MEDLINE | ID: mdl-29442453

BACKGROUND: Arsenic trioxide (ATO) is a well-recognized antileukemic drug used for the treatment of newly diagnosed and relapsed acute promyelocytic leukemia (APL). A major drawback of therapy with ATO is the development of APL cell resistance, the mechanisms of which are still not clear. OBJECTIVES: The aim of this study was to investigate the role of the PI3K/Akt signaling pathway in ATOtreated human acute myeloid leukemia (HL-60) cells and in ATO-resistant clones. MATERIAL AND METHODS: The cytotoxicity of ATO was assessed using Trypan blue staining or a WST-1 reduction assay. The Akt phosphorylation level was measured by immunofluorescent staining and flow cytometry. Gene expression analysis was performed using real-time polymerase chain reaction (PCR). RESULTS: The clones derived by culturing for 8-12 weeks in the presence of 1.75, 2.5, and 5 µM ATO were characterized by high viability but a slower growth rate compared to the parental HL-60 cells. The flow cytometry analysis showed that in the parental cells the levels of p-Akt were undetectable or very low, and that ATO had no effect on the level of p-Akt in either the ATO-treated parental cells or the clones. The gene expression analysis revealed that some of the genes involved in the Akt pathway may play a key role in the induction of resistance to ATO, e.g., genes encoding cyclin D1 (CCND1), fork head box O1 (FOXO1), Jun oncogene (JUN), protein kinase C isoform B1 (PRKCB1), because their expression profiles were predominantly changed in the clones and/or the ATO-treated parental HL-60 cells. CONCLUSIONS: The overall results indicate that CCND1, FOXO1, and JUN may contribute to the induction of resistance to ATO, and that the C-Jun N-terminal kinase (JNK) signaling pathway may have greater significance than the phosphoinositide 3-kinase (PI3K)/Akt pathway in mediating the cytotoxic effects of ATO and the development of resistance to ATO in the HL-60 cell line.


Antineoplastic Agents/pharmacology , Gene Expression Profiling , Genes, jun/physiology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , Arsenic Trioxide , Arsenicals , Cyclin D1/physiology , Drug Resistance, Neoplasm , Forkhead Box Protein O1/physiology , HL-60 Cells , Humans , Oxides , Protein Kinase C beta/physiology
17.
Leukemia ; 31(8): 1735-1742, 2017 08.
Article En | MEDLINE | ID: mdl-28025584

Immunoglobulin light chain (AL) amyloidosis is characterized by tissue deposition of amyloid fibers derived from immunoglobulin light chain. AL amyloidosis and multiple myeloma (MM) originate from monoclonal gammopathy of undetermined significance. We wanted to characterize germline susceptibility to AL amyloidosis using a genome-wide association study (GWAS) on 1229 AL amyloidosis patients from Germany, UK and Italy, and 7526 healthy local controls. For comparison with MM, recent GWAS data on 3790 cases were used. For AL amyloidosis, single nucleotide polymorphisms (SNPs) at 10 loci showed evidence of an association at P<10-5 with homogeneity of results from the 3 sample sets; some of these were previously documented to influence MM risk, including the SNP at the IRF4 binding site. In AL amyloidosis, rs9344 at the splice site of cyclin D1, promoting translocation (11;14), reached the highest significance, P=7.80 × 10-11; the SNP was only marginally significant in MM. SNP rs79419269 close to gene SMARCD3 involved in chromatin remodeling was also significant (P=5.2 × 10-8). These data provide evidence for common genetic susceptibility to AL amyloidosis and MM. Cyclin D1 is a more prominent driver in AL amyloidosis than in MM, but the links to aggregation of light chains need to be demonstrated.


Amyloidosis/genetics , Genome-Wide Association Study , Immunoglobulin Light Chains/metabolism , Multiple Myeloma/genetics , Adult , Aged , Aged, 80 and over , Cyclin D1/physiology , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide
18.
Oncotarget ; 7(29): 45214-45224, 2016 07 19.
Article En | MEDLINE | ID: mdl-27286258

The interactions of multiple myeloma (MM) cells with their microenvironment are crucial for pathogenesis. MM cells could interact differentially with their microenvironment depending on the type of cyclin D they express. We established several clones that constitutively express cyclin D1 from the parental RPMI8226 MM cell line and analyzed the impact of cyclin D1 expression on cell behavior. We performed a gene expression profiling study on cyclin D1-expressing vs. control cells and validated the results by semi-quantitative RT-PCR. The expression of cyclin D1 altered the transcription of genes that control adhesion and migration. We confirmed that cyclin D1 increases cell adhesion to stromal cells and fibronectin, stabilizes F-actin fibers, and enhances chemotaxis and inflammatory chemokine secretion. Both control and cyclin D1-expressing cells were more resistant to acute carfilzomib treatment when cultured on stromal cells than in suspension. However, this resistance was specifically reduced in cyclin D1-expressing cells after pomalidomide pre-treatment that modifies tumor cell/microenvironment interactions. Transcriptomic analysis revealed that cyclin D1 expression was also associated with changes in the expression of genes controlling metabolism. We also found that cyclin D1 expression disrupted the redox balance by producing reactive oxygen species. The resulting oxidative stress activated the p44/42 mitogen-activated protein kinase (or ERK1/2) signaling pathway, increased cell adhesion to fibronectin or stromal cells, and controlled drug sensitivity.Our results have uncovered a new function for cyclin D1 in the control of redox metabolism and interactions of cyclin D1-expressing MM cells with their bone marrow microenvironment.


Cyclin D1/physiology , Multiple Myeloma/pathology , Cell Adhesion , Cell Line, Tumor , Cell Movement , Chemokines/metabolism , Cyclin D1/genetics , Drug Resistance, Neoplasm , Extracellular Signal-Regulated MAP Kinases/physiology , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species/metabolism , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Tumor Microenvironment
19.
J Diabetes Investig ; 7(1): 27-8, 2016 Jan.
Article En | MEDLINE | ID: mdl-26816597

Dysregulation of gluconeogenesis is a key pathological feature of type 2 diabetes. However, the molecular mechanisms underlying the regulation of gluconeogenesis remain unclear. Bhalla et al. recently reported that cyclin D1 suppresses hepatic gluconeogenesis through CDK4-dependent phosphorylation of PGC1alpha and consequent inhibition of its activity. The cyclin D1-CDK4 might thus serve as an important link between the cell cycle and control of energy metabolism through modulation of PGC1alpha activity.


Cell Cycle/physiology , Cyclin D1/physiology , Cyclin-Dependent Kinase 4/physiology , Gluconeogenesis/physiology , Animals , Humans
20.
Tumour Biol ; 37(6): 7395-404, 2016 Jun.
Article En | MEDLINE | ID: mdl-26676638

The Forkhead box P3 (FOXP3) transcription factor is the key driver of the differentiation and immunosuppressive function of regulatory T cells (Tregs). Additionally, FOXP3 has been reported to be expressed in many solid tumor cell lines and tissues. However, its role in tumorigenesis and tumor progression is conflicting, both tumor suppressive and promoting functions have been described. In this study, we demonstrated that FOXP3 was expressed in both lung adenocarcinoma tissues and the lung adenocarcinoma cell line A549. FOXP3 inhibition decreased cell proliferation, migration, and invasion as well as the secretion of inhibitory cytokines (e.g., transforming growth factor beta 1 (TGF-ß1), interleukin 35 (IL-35), and heme oxygenase-1 (HMOX1)), suggesting a positive role for FOXP3 in tumor development. Importantly, we found that FOXP3 could enhance lung adenocarcinoma cell proliferation via upregulating the levels of the cell cycle G1/S checkpoint gene CCND1. These data demonstrated that FOXP3 could be regarded as a novel therapeutic target for inhibiting lung adenocarcinoma progression.


Adenocarcinoma/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Cyclin D1/physiology , Forkhead Transcription Factors/physiology , G1 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Neoplasm Proteins/physiology , A549 Cells , Adenocarcinoma/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Division , Cyclin D1/biosynthesis , Cyclin D1/genetics , Cytokines/metabolism , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Humans , Lung Neoplasms/pathology , Neoplasm Invasiveness , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , RNA Interference , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , RNA, Small Interfering/genetics , Tumor Escape/genetics , Tumor Escape/physiology , Up-Regulation
...