Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Int J Biol Sci ; 20(1): 387-402, 2024.
Article in English | MEDLINE | ID: mdl-38164176

ABSTRACT

Colon adenocarcinoma (COAD) is the most common malignancy of the digestive tract, which is characterized by a dismal prognosis. No effective treatment has been established presently, thus there is an urgent need to understand the mechanisms driving COAD progression in order to develop effective therapeutic approaches and enhance clinical outcomes. In this study, we found that KLF7 is overexpressed in COAD tissues and correlated with clinicopathological features of COAD. Both gain-of-function and loss-of-function experiments have unequivocally demonstrated that overexpression of KLF7 promotes the growth and metastasis of COAD in vitro and in vivo, while KLF7 knockdown attenuated these effects. Mechanistically, our findings reveal that KLF7 can specifically bind to the promoter region of PDGFB (TGGGTGGAG), thus promoting the transcription of PDGFB and increasing its secretion. Subsequently, secreted PDGFB facilitates the progression of COAD by activating MAPK/ERK, PI3K/AKT, and JAK/STAT3 signaling pathways through PDGFRß. Additionally, we found that sunitinib can block PDGFB signaling and inhibit COAD progression, offering a promising therapeutic strategy for COAD treatment.


Subject(s)
Adenocarcinoma , Colonic Neoplasms , Humans , Colonic Neoplasms/metabolism , Proto-Oncogene Proteins c-sis/metabolism , Adenocarcinoma/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/genetics , Becaplermin , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism
2.
Pharmaceutics ; 15(9)2023 Sep 04.
Article in English | MEDLINE | ID: mdl-37765244

ABSTRACT

Sonodynamic therapy (SDT) induces reactive oxygen species (ROS) to kill tumor cells. Heme oxygenase-1 (HO-1), as an important antioxidant enzyme, resists killing by scavenging ROS. Zinc protoporphyrin (ZnPP) not only effectively inhibits HO-1 activity, but also becomes a potential sonosensitizer. However, its poor water solubility limits its applications. Herein, we developed an improved water-soluble method. It was proved that pegylated zinc protoporphyrin-mediated SDT (PEG-ZnPP-SDT) could significantly enhance ROS production by destroying the HO-1 antioxidant system in ovarian cancer. Increased ROS could cause mitochondrial membrane potential collapse, release cytochrome c from mitochondria to the cytoplasm, and trigger the mitochondrial-caspase apoptotic pathway. In conclusion, our results demonstrated that PEG-ZnPP-SDT, as a novel sonosensitizer, could improve the antitumor effects by destroying the HO-1 antioxidant system. It provided a new therapeutic strategy for SDT to treat cancers, especially those with higher HO-1 expression.

3.
Int J Mol Sci ; 23(18)2022 Sep 12.
Article in English | MEDLINE | ID: mdl-36142474

ABSTRACT

Photodynamic therapy (PDT) has significant advantages in the treatment of malignant tumors, such as high efficiency, minimal invasion and less side effects, and it can preserve the integrity and quality of the organs. The power density, irradiation time and photosensitizer (PS) concentration are three main parameters that play important roles in killing tumor cells. However, until now, the underlying relationships among them for PDT outcomes have been unclear. In this study, human malignant glioblastoma U-118MG and melanoma A375 cells were selected, and the product of the power density, irradiation time and PS concentration was defined as the total photodynamic parameter (TPP), in order to investigate the mechanisms of PS sinoporphyrin sodium (DVDMS)-mediated PDT (DVDMS-PDT). The results showed that the survival rates of the U-118MG and A375 cells were negatively correlated with the TPP value in the curve, and the correlation exactly filed an e-exponential function. Moreover, according to the formula, we realized controllable killing effects of the tumor cells by randomly adjusting the three parameters, and we finally verified the accuracy and repeatability of the formula. In conclusion, the establishment and implementation of a newly functional relationship among the PDT parameters are essential for predicting PDT outcomes and providing personalized precise treatment, and they are contributive to the development of PDT dosimetry.


Subject(s)
Photochemotherapy , Porphyrins , Cell Line, Tumor , Humans , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Porphyrins/pharmacology , Porphyrins/therapeutic use
4.
Free Radic Res ; 55(9-10): 958-969, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34670466

ABSTRACT

Sonodynamic therapy (SDT) represents a noninvasive therapeutic method via the activation of certain chemical sensitizers using low-intensity ultrasound to generate various reactive oxygen species (ROS). In this work, we conducted systematic experiments to evaluate the production of hydrogen peroxide (H2O2) in sinoporphyrin sodium (DVDMS) mediated SDT (DVDMS-SDT). We found that the fluorescence intensities of H2O2-specific probe BES-H2O2 and Amplex Red increased significantly exposure to DVDMS-SDT while decreased with the introduction of catalase (H2O2 scavenger), indicating the production of H2O2. And the fluorescence intensity of H2O2 susceptible probes were positively correlated with DVDMS concentration, ultrasound intensity and irradiation time. Under the same molarity concentration, DVDMS has advantages over proto-porphyrin IX (PpIX) and hemoporrin monomethyl ether (HMME) in H2O2 production, indicating that the yield of H2O2 depends on the properties of sensitizer. More importantly, DVDMS-SDT is involved in the process of H2O2 even in the oxygen-free condition, showing its greater superiority for the treatment of tumor under hypoxia environment.


Subject(s)
Porphyrins , Ultrasonic Therapy , Cell Line, Tumor , Humans , Hydrogen Peroxide , Hypoxia , Porphyrins/chemistry , Porphyrins/therapeutic use , Reactive Oxygen Species , Ultrasonic Therapy/methods
5.
Cell Death Dis ; 11(9): 792, 2020 09 23.
Article in English | MEDLINE | ID: mdl-32968053

ABSTRACT

Cytoplasmic polyadenylation element-binding protein 3 (CPEB3) is a sequence-specific RNA-binding protein. We had reported that CPEB3 is involved in hepatocellular carcinoma (HCC) progression. However, the underlying mechanisms of CPEB3 in HCC remain unclear. In this study, we firstly performed RNA immunoprecipitation to uncover the transcriptome-wide CPEB3-bound mRNAs (CPEB3 binder) in HCC. Bioinformatic analysis indicates that CPEB3 binders are closely related to cancer progression, especially HCC metastasis. Further studies confirmed that metadherin (MTDH) is a direct target of CPEB3. CPEB3 can suppress the translation of MTDH mRNA in vivo and in vitro. Besides, luciferase assay demonstrated that CPEB3 interacted with 3'-untranslated region of MTDH mRNA and inhibited its translation. Subsequently, CPEB3 inhibited the epithelial-mesenchymal transition and metastasis of HCC cells through post-transcriptional regulation of MTDH. In addition, cpeb3 knockout mice are more susceptible to carcinogen-induced hepatocarcinogenesis and subsequent lung metastasis. Our results also indicated that CPEB3 was a good prognosis marker, which is downregulated in HCC tissue. In conclusion, our results demonstrated that CPEB3 played an important role in HCC progression and targeting CPEB3-mediated mRNA translation might be a favorable therapeutic approach.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Membrane Proteins/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Carcinogenesis/genetics , Carcinoma, Hepatocellular/metabolism , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Liver Neoplasms/metabolism , Membrane Proteins/metabolism
6.
Int J Biochem Cell Biol ; 113: 104-114, 2019 08.
Article in English | MEDLINE | ID: mdl-30660690

ABSTRACT

Sonodynamic therapy (SDT) is a noninvasive therapeutic method via the activation of certain chemical sensitizers using low intensity ultrasound. In this work, we evaluated the antitumor effect of sinoporphyrin sodium (DVDMS) mediated SDT (DVDMS-SDT) on Hepatocellular carcinoma (HCC) cell lines both in vitro and in vivo. The results indicated that DVDMS-SDT was significantly more efficacious than PpIX-SDT in treating hepatocellular cell line Hep-G2. DVDMS-SDT also increased the ratio of cells in the G2/M phase and decreased the CDK1 and Cyclin B1 protein level. DVDMS-SDT markedly increased intracellular reactive oxygen species (ROS) in vitro. The increased ROS production up-regulated the expression of p53 and Bax, and down-regulated Bcl-2 expression, which led to the activation of caspase-3, ultimately initiated cell apoptosis. These effects could be partially reversed by the ROS scavenger N-acetylcysteine (NAC). In vivo experiments revealed that the DVDMS-SDT resulted in an effective inhibition of tumor growth and prolonged the survival time of tumor-bearing mice. More importantly, no obvious signs of side effects were observed. These results suggested that DVDMS-SDT is very effective in treating Hepatocellular carcinoma without side effects. The primary mechanism of SDT is due to the increased ROS activated the p53/Caspase 3 axis of apoptosis.


Subject(s)
Carcinoma, Hepatocellular/therapy , Caspase 3/metabolism , Liver Neoplasms/therapy , Porphyrins/pharmacology , Tumor Suppressor Protein p53/metabolism , Ultrasonic Therapy , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Female , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
7.
Int J Oncol ; 53(4): 1691-1702, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30066913

ABSTRACT

MicroRNAs (miRNAs or miRs) play important roles in numerous cellular processes, including development, proliferation, tumorigenesis and apoptosis. It has been reported that miRNA expression is induced by ionizing radiation (IR) in cancer cells. However, the underlying molecular mechanisms are not yet fully understood. In this study, endogenous miR­320a and its primary precursor (pri­miR­320a) were assayed by reverse transcription­quantitative PCR (RT­qPCR). Luciferase activities were measured using a dual­luciferase reporter assay system. Western blot analysis was used to determine the protein expressions of upstream and downstream genes of miR­320a. Cell apoptosis was evaluated by Annexin V apoptosis assay and cell proliferation was measured using the trypan blue exclusion method. The results revealed that miR­320a expression increased linearly with the IR dose and treatment duration. Three transcription factors, activating transcription factor 2 (ATF2), ETS transcription factor (ELK1) and YY1 transcription factor (YY1), were activated by p38 mitogen­activated protein kinase (MAPK) and mitogen­activated protein kinase 8 (JNK) and by upregulated miR­320a expression under IR conditions. In addition, it was identified that X­linked inhibitor of apoptosis (XIAP) was an miR­320a target gene during the IR response. By targeting XIAP, miR­320a induced apoptosis and inhibited the proliferation of the cancer cells. On the whole, the results of this study demonstrated that miRNA­320a, regulated by the p38 MAPK/JNK pathway, enhanced the radiosensitivity of cancer cells by inhibiting XIAP and this may thus prove to be a potential therapeutic approach with which to overcome radioresistance in cancer treatment.


Subject(s)
Apoptosis/radiation effects , MAP Kinase Signaling System/radiation effects , MicroRNAs/genetics , Neoplasms/radiotherapy , X-Linked Inhibitor of Apoptosis Protein/genetics , Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/metabolism , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/radiation effects , Dose-Response Relationship, Radiation , Gene Expression Regulation, Neoplastic/radiation effects , Humans , MAP Kinase Signaling System/genetics , MicroRNAs/metabolism , Neoplasms/genetics , Promoter Regions, Genetic , RNA, Small Interfering/metabolism , Radiation Tolerance/genetics , Radiation, Ionizing , Treatment Outcome , Up-Regulation/radiation effects , X-Linked Inhibitor of Apoptosis Protein/metabolism , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism , ets-Domain Protein Elk-1/genetics , ets-Domain Protein Elk-1/metabolism
8.
J Exp Clin Cancer Res ; 37(1): 46, 2018 Mar 05.
Article in English | MEDLINE | ID: mdl-29506532

ABSTRACT

BACKGROUND: Cell adhesion molecules (CADMs) comprise of a protein family whose functions include maintenance of cell polarity and tumor suppression. Hypo-expression of CADM2 gene expression has been observed in several cancers including hepatocellular carcinoma (HCC). However, the role and mechanisms of CADM2 in HCC remain unclear. METHODS: The expression of CADM2 and miRNA-10b (miR-10b) in HCC tissues and cell lines were detected using real-time PCR and Western blotting. Immunofluorescence was used to detect Epithelial-mesenchymal transition (EMT) progression in HCC cell lines. Dual-luciferase reporter assay was used to determine miR-10b binding to CADM2 3'UTR. Wound healing assay and Transwell assay were performed to examine the migration and invasion of HCC cells. RESULTS: We report the effect of CADM2 as a tumor suppressor in HCC. Firstly, we confirmed that CADM2 expression was significantly down regulated in HCC tissues compared to normal tissues according to TCGA data analysis and fresh HCC sample detection. Secondly, overexpression of CADM2 could inhibit EMT process, migratory and invasion ability of HCC cells. Furthermore, the results indicated that CADM2 is a direct target of miR-10b in HCC cells and miR-10b/CADM2 modulates EMT process and migration ability via focal adhesion kinase (FAK) /AKT signaling pathway in HCC. CONCLUSIONS: Our study demonstrates that miR-10b-CADM2-FAK/AKT axis plays an important role in HCC metastasis, which might be a novel potential therapeutic option for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Adhesion Molecules/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , 3' Untranslated Regions , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cell Adhesion Molecules/metabolism , Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression , Genes, Reporter , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Models, Biological , Neoplasm Metastasis , Neoplasm Staging , Prognosis , RNA Interference
10.
J Lipid Res ; 58(8): 1548-1560, 2017 08.
Article in English | MEDLINE | ID: mdl-28536109

ABSTRACT

Both iron and lipids are involved in the progression of alcoholic fatty liver disease (AFLD), but the interaction between iron and lipids in AFLD is unclear. Here, we tested the hypothesis that iron regulates the expression of genes involved in lipid metabolism through iron regulatory proteins (IRPs), which interact with the iron-responsive elements (IREs) in the untranslated regions (UTRs) of genes, resulting in lipid accumulation. Using "RNA structure software", we predicted the mRNA secondary structures of more than 100 genes involved in lipid metabolism to investigate whether the IRE structure exists in novel mRNAs. Cholesterol 7α-hydroxylase (Cyp7a1) has an IRE-like stem-loop, a noncanonical IRE structure, in its 3'-UTR. Cyp7a1 expression can be regulated by in vivo and in vitro iron treatment. In addition, the noncanonical IRE motif can efficiently bind both to IRP1 and IRP2. The results indicate that hepatic iron overloading in AFLD mice decreased Cyp7a1 expression and resulted in cholesterol accumulation, providing a new mechanism of iron-regulated gene transcription and translation through the interaction between iron and a noncanonical IRE structure in Cyp7a1 mRNA. This finding has significant implications in studying a proposed mechanism for the regulation of cholesterol homeostasis by an Fe/IRP/noncanonical IRE axis.


Subject(s)
Cholesterol 7-alpha-Hydroxylase/genetics , Ethanol/adverse effects , Fatty Liver, Alcoholic/genetics , Gene Expression Regulation, Enzymologic/drug effects , Iron/pharmacology , 3' Untranslated Regions/genetics , Animals , Base Sequence , Cell Line , Fatty Liver, Alcoholic/metabolism , Mice , Mice, Inbred C57BL , RNA Stability , Response Elements/genetics
11.
Liver Int ; 37(9): 1354-1364, 2017 09.
Article in English | MEDLINE | ID: mdl-28317284

ABSTRACT

BACKGROUND & AIMS: Several studies have shown that miR-320a induces apoptosis, inhibits cell proliferation, and affects cell cycle progression as a tumour suppressor in many cancers. However, the involvement of miR-320a in the invasion and metastasis of hepatocellular carcinoma (HCC) is still unknown. METHODS: Endogenous miR-320a and high mobility group box 1 (HMGB1) expressions were assayed by real-time PCR. Luciferase activities were measured using a dual-luciferase reporter assay system. Western blots were used to determine the protein expressions of HMGB1, MMP2, and MMP9. Invasion and metastasis of tumour cells were, respectively, evaluated by the transwell invasion assay and the wound healing assay. RESULTS: The expression of miR-320a was significantly decreased in 24 of 32 (75%) HCC tissues and associated with the invasion and metastasis of HCC. Furthermore, we demonstrated that HMGB1 was a direct target of miR-320a and there was a significant negative correlation between miR-320a and HMGB1 expression in HCC. Ectopic expression or inhibition of miR-320a potently regulated the invasion and metastasis of HCC cells in HMGB1-dependent manner. CONCLUSIONS: Our results showed that miR-320a was involved in the invasion and metastasis by targeting HMGB1 and had an anti-metastasis effect in HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic , HMGB1 Protein/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Adult , Aged , Apoptosis , Carcinoma, Hepatocellular/pathology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Disease Progression , Female , HMGB1 Protein/metabolism , Humans , Liver Neoplasms/pathology , Male , MicroRNAs/metabolism , Middle Aged , Neoplasm Invasiveness
12.
J Exp Clin Cancer Res ; 35: 71, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-27102814

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) accounts for 75% of liver cancers and is the second most lethal cancer, associated with its multiple etiologies, poor prognosis and resistance to chemotherapy drugs. Chemotherapy treatment on HCC suffers low efficacy of drug uptake and can produce a range of side effects. Here we report an investigation on the effect of a combined treatment on human hepatocellular carcinoma BEL-7402 cells using low-intensity ultrasound (US) and 5-fluorouracil (5-FU). METHODS: The uptake of 5-FU was measured by the high-performance liquid chromatography (HPLC). DNA damage was detected by the comet assay. MTT assay was used to examine cell viability. Intracellular reactive oxygen species (ROS) and mitochondrial membrane potential (Δψm) were respectively detected by the fluorescent probes DCFH-DA or JC-1. Endogenous apoptosis-associated proteins were analyzed by the western blot and immunohistochemistry. Histopathological changes were evaluated by the hematoxylin and eosin (H&E) staining. Cell apoptosis was evaluated by the TUNEL and flow cytometry assays. Cell proliferation was measured using the immunohistochemical staining of PCNA. RESULTS: Our results showed that low-intensity US (1.1 MHz, 1.0 W/cm2, 10% duty cycle) significantly enhanced the uptake of 5-FU, 5-FU-mediated DNA damage and reactive oxygen species (ROS) generation. The increased ROS production up-regulated the p53 protein level, which led to the up-regulation of Bax and down-regulation of Bcl-2. The enhancement of ROS generation and the activation of the apoptosis-associated proteins further triggered the collapse of mitochondrial membrane potential, released cytochrome c from mitochondria into cytosol and activated the mitochondria-caspase pathway, and cell apoptosis. Such enhanced effects could be partially blocked by the ROS scavenger N-acetylcysteine (NAC). Overall, low-intensity US combined with 5-FU led to an effective inhibition of tumor growth and prolonged overall survival of BEL-7402 HCC-bearing nude mice by more than 15% compared with 5-FU treatment alone. CONCLUSIONS: Our results showed that low-intensity ultrasound combined with 5-FU produced much enhanced synergistic anti-tumor effects via enhanced ROS production in treating HCC.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Carcinoma, Hepatocellular/therapy , Fluorouracil/administration & dosage , Liver Neoplasms/therapy , Ultrasonic Therapy/methods , Animals , Antimetabolites, Antineoplastic/pharmacology , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Combined Modality Therapy , Down-Regulation , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Reactive Oxygen Species/metabolism , Treatment Outcome , Up-Regulation , Xenograft Model Antitumor Assays
13.
Oncotarget ; 7(16): 22159-73, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-26989025

ABSTRACT

The overall biological role and clinical significance of long non-coding RNA H19 in colorectal cancer (CRC) remain largely unknown. Here, we firstly report that the lncRNA H19 recruits eIF4A3 and promotes the CRC cell proliferation. We observed higher expression of H19 was significantly correlated with tumor differentiation and advanced TNM stage in a cohort of 83 CRC patients. Multivariate analyses revealed that expression of H19 served as an independent predictor for overall survival and disease-free survival. Further experiments revealed that overexpression of H19 promoted the proliferation of CRC cells, while depletion of H19 inhibited cell viability and induced growth arrest. Moreover, expression profile data showed that H19 upregulated a series of cell-cycle genes. Using bioinformatics prediction and RNA immunoprecipitation assays, we identified eIF4A3 as an RNA-binding protein that binds to H19. We confirmed that combining eIF4A3 with H19 obstructed the recruitment of eIF4A3 to the cell-cycle gene mRNA. Our results suggest that H19, as a growth regulator, could serve as a candidate prognostic biomarker and target for new therapies in human CRC.


Subject(s)
Adenocarcinoma/pathology , Colorectal Neoplasms/pathology , DEAD-box RNA Helicases/metabolism , Eukaryotic Initiation Factor-4A/metabolism , Gene Expression Regulation, Neoplastic/genetics , RNA, Long Noncoding/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adult , Aged , Cell Proliferation/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Proportional Hazards Models
14.
PLoS One ; 11(1): e0147084, 2016.
Article in English | MEDLINE | ID: mdl-26765329

ABSTRACT

Renal fibrosis plays an important role in the onset and progression of chronic kidney diseases. Many studies have demonstrated that heme oxygenase-1 (HO-1) is involved in diverse biological processes as a cytoprotective molecule, including anti-inflammatory, anti-oxidant, anti-apoptotic, antiproliferative, and immunomodulatory effects. However, the mechanisms of HO-1 prevention in renal interstitial fibrosis remain unknown. In this study, HO-1 transgenic (TG) mice were employed to investigate the effect of HO-1 on renal fibrosis using a unilateral ureter obstruction (UUO) model and to explore the potential mechanisms. We found that HO-1 was adaptively upregulated in kidneys of both TG and wild type (WT) mice after UUO. The levels of HO-1 mRNA and protein were increased in TG mice compared with WT mice under normal conditions. HO-1 expression was further enhanced after UUO and remained high during the entire experimental process. Renal interstitial fibrosis in the TG group was significantly attenuated compared with that in the WT group after UUO. Moreover, overexpression of HO-1 inhibited the loss of peritubular capillaries. In addition, UUO-induced activation and proliferation of myofibroblasts were suppressed by HO-1 overexpression. Furthermore, HO-1 restrained tubulointerstitial infiltration of macrophages and regulated the secretion of inflammatory cytokines in UUO mice. We also found that high expression of HO-1 inhibited reactivation of Wnt/ß-catenin signaling, which could play a crucial role in attenuating renal fibrosis. In conclusion, these data suggest that HO-1 prevents renal tubulointerstitial fibrosis possibly by regulating the inflammatory response and Wnt/ß-catenin signaling. This study provides evidence that augmentation of HO-1 levels may be a therapeutic strategy against renal interstitial fibrosis.


Subject(s)
Gene Expression , Heme Oxygenase-1/genetics , Nephritis, Interstitial/etiology , Nephritis, Interstitial/pathology , Ureteral Obstruction/complications , Animals , Apoptosis/genetics , Cell Proliferation , Disease Models, Animal , Fibrosis , Gene Expression Regulation , Mice , Myofibroblasts/metabolism , Up-Regulation , Wnt Signaling Pathway
15.
J Dermatol Sci ; 79(2): 155-62, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25982144

ABSTRACT

BACKGROUD: Malignant melanoma is a very refractory skin tumor due to its high metastasis, poor prognosis, and insensitivity to chemotherapy. Sonodynamic therapy has recently evolved as a potential method to treat cancers. In this study, 5-aminolevulinic acid-mediated sonodynamic therapy (ALA-SDT) was used to treat malignant melanoma in vivo. OBJECTIVE: To investigate whether ALA-SDT induces anti-tumor effects in malignant melanoma and to see if miRNAs are involved in this process. METHODS: Tumor transplantation experiments in BALB/c nude mice were used to assess anti-tumor effects after ALA-SDT treatment. Cell apoptosis was evaluated by TUNEL assays and cell proliferation was measured using immunohistochemisty with anti-PCNA antibody. Microarray analysis was performed to measure miRNAs expressions. Endogenous miR-34a and its upstream and downstream genes were assayed by real-time PCR. Western blottings were used to determine these protein expressions. Intracellular ROS levels were detected by measuring the fluorescence intensity of DCF. RESULTS: Tumor transplantation experiments revealed that ALA-SDT could inhibit mouse melanoma cell proliferation and tumor growth. Compared with the control group, TUNEL assays revealed that apoptosis was increased and proliferation was inhibited in the SDT group. Real-time PCR analysis showed 14-fold increase of miR-34a expression in the SDT group compared to the control group. In addition, ALA-SDT significantly increased intracellular ROS levels in vitro, which were almost inhibited by the ROS scavenger NAC. Also, the mRNA, total protein, and acetylation levels of p53 were increased, whereas some downstream anti-apoptotic or pro-proliferative factors of miR-34a such as BCL2, CCND1, CDK6, and SIRT1 were decreased in the SDT group compared with the control, ALA alone, and ultrasound alone groups. When miR-34a was inhibited in vitro, the protein expressions of BCL2, CCND1, CDK6, and SIRT1 recovered. By targeting SIRT1, which inhibits p53 acetylation, miR-34a promoted the transcriptional activity of p53, and finally led to increased expression of miR-34a itself. Therefore, the p53, miR-34a, and SIRT1 constituted a positive feedback loop. CONCLUSION: ALA-SDT showed synergistic anti-tumor effects in malignant melanoma by constituting a positive feedback loop of p53-miR-34a-Sirt1 axis.


Subject(s)
Aminolevulinic Acid/therapeutic use , Melanoma, Experimental/drug therapy , MicroRNAs/metabolism , Photosensitizing Agents/therapeutic use , Sirtuin 1/metabolism , Tumor Suppressor Protein p53/metabolism , Ultrasonic Therapy , Animals , Cell Line, Tumor , Elasticity Imaging Techniques , Melanoma, Experimental/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Animal , Photochemotherapy/methods , Real-Time Polymerase Chain Reaction
16.
Mol Cell Biol ; 35(6): 988-1000, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25582198

ABSTRACT

MicroRNAs (miRNAs) are dysregulated in many types of malignant diseases, including colorectal cancer. miRNA 30a (miR-30a) is a member of the miR-30 family and has been implicated in many types of cancers. In this study, we determined the expression of miR-30a in human colon cancer tissues and cell lines. miR-30a was found to be significantly downregulated in both the tissues and cell lines. Furthermore, overexpression of miR-30a inhibited, while silencing of miR-30a promoted, cell proliferation, migration, and invasion in vitro. Consistently, stable overexpression of miR-30a suppressed the growth of colon cancer cell xenografts in vivo. Moreover, bioinformatic algorithms and luciferase reporter assays revealed that insulin receptor substrate 2 (IRS2) is a direct target of miR-30a. Further functional studies suggested that repression of IRS2 by miR-30a partially mediated the tumor suppressor effect of miR-30a. In addition, miR-30a inhibited constitutive phosphorylation of Akt by targeting IRS2. Additionally, clinicopathological analysis indicated that miR-30a has an inverse correlation with the staging in patients with colon cancer. Taken together, our study provides the first evidence that miR-30a suppressed colon cancer cell growth through inhibition of IRS2. Thus, miR-30a might serve as a promising therapeutic strategy for colon cancer treatment.


Subject(s)
Carcinogenesis/genetics , Colorectal Neoplasms/genetics , Insulin Receptor Substrate Proteins/genetics , MicroRNAs/genetics , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Down-Regulation/genetics , Female , Genes, Tumor Suppressor , HCT116 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Proto-Oncogene Proteins c-akt/genetics
17.
Oncol Rep ; 32(3): 1021-8, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24968707

ABSTRACT

Accumulating evidence suggests that microRNAs (miRNAs) can function as oncogenes or as tumor suppressor genes depending on the tissue type or target. Therefore, clarification of the specific roles of miRNAs is vital for the diagnosis and treatment of cancer. In the present study, miR-451 was found to be downregulated in hepatocellular carcinoma (HCC) tissues when compared to that in adjacent tissues. Functional analysis showed that, in vitro, miR-451 inhibited the migration of hepatoma cell lines HepG2 and SK-Hep-1. Further investigation of the molecular mechanisms identified activating transcription factor 2 (ATF2) as a target of miR-451. miR-451 inhibited ATF2 expression by binding to the 3'UTR. An in vivo assay revealed a significant negative correlation between miR-451 and ATF2 in liver cancer tissues. According to previous findings reported in the literature, the opposing functions of ATF2 are related to its subcellular localization. In the nucleus, ATF2 displays oncogenic activities in melanoma. In the present study, ATF2 exhibited a higher expression level in the nucleus in tumoral tissues of HCC as detected by immunohistochemistry. In conclusion, in this study, we identified a potential target of miR-451, ATF2, and revealed a novel role of miR-451 in the inhibition of the migratory ability of hepatoma cell lines.


Subject(s)
Activating Transcription Factor 2/genetics , Carcinoma, Hepatocellular/genetics , Cell Nucleus/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Adult , Aged , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement , Down-Regulation , Female , Hep G2 Cells , Humans , Liver Neoplasms/pathology , Male , Middle Aged
18.
J Cell Mol Med ; 18(6): 966-74, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24801603

ABSTRACT

The transition from liver fibrosis to hepatocellular carcinoma (HCC) has been suggested to be a continuous and developmental pathological process. MicroRNAs (miRNAs) are recently discovered molecules that regulate the expression of genes involved in liver disease. Many reports demonstrate that miR-483-5p and miR-483-3p, which originate from miR-483, are up-regulated in HCC, and their oncogenic targets have been identified. However, recent studies have suggested that miR-483-5p/3p is partially down-regulated in HCC samples and is down-regulated in rat liver fibrosis. Therefore, the aberrant expression and function of miR-483 in liver fibrosis remains elusive. In this study, we demonstrate that overexpression of miR-483 in vivo inhibits mouse liver fibrosis induced by CCl4 . We demonstrate that miR-483-5p/3p acts together to target two pro-fibrosis factors, platelet-derived growth factor-ß and tissue inhibitor of metalloproteinase 2, which suppress the activation of hepatic stellate cells (HSC) LX-2. Our work identifies the pathway that regulates liver fibrosis by inhibiting the activation of HSCs.


Subject(s)
Carcinoma, Hepatocellular/prevention & control , Hepatic Stellate Cells/cytology , Liver Cirrhosis/prevention & control , MicroRNAs/genetics , Platelet-Derived Growth Factor/antagonists & inhibitors , Tissue Inhibitor of Metalloproteinase-2/antagonists & inhibitors , Transforming Growth Factor beta/pharmacology , Animals , Carbon Tetrachloride/toxicity , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/metabolism , Cells, Cultured , Coculture Techniques , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Humans , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Neoplasms/etiology , Liver Neoplasms/metabolism , Liver Neoplasms/prevention & control , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/metabolism , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , RNA, Messenger/genetics , Rats , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism
19.
FEBS Lett ; 585(19): 3095-100, 2011 Oct 03.
Article in English | MEDLINE | ID: mdl-21893058

ABSTRACT

Angiogenesis, a key factor in ischemic heart disease, is rapidly initiated in response to hypoxic or ischemic conditions. MicroRNAs (miRNAs) are endogenously expressed small non-coding RNAs that regulate gene expression at post-transcriptional level. The recent discovery of the involvement of these RNAs in the control of angiogenesis renders them very attractive in the development of new approaches for restoring the angiogenic balance. In the present study, we explored that miR-483-5p, a microRNA embedded in the intron of insulin-like growth factor 2 (Igf2), acts as an endogenous angiogenesis-inhibiting factor. We identified that serum response factor (SRF) is one of miR-483-5p target genes. These findings indicated that the miR-483-5p-SRF pathway may offer a novel strategy for treatment with angiogenesis in ischemic heart disease patients.


Subject(s)
MicroRNAs/metabolism , Neovascularization, Physiologic/physiology , Serum Response Factor/metabolism , 3' Untranslated Regions , Cell Line , Endothelial Cells/cytology , Endothelial Cells/physiology , Gene Expression Regulation , Humans , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Introns , MicroRNAs/genetics , Serum Response Factor/genetics , Signal Transduction/physiology
20.
Anat Rec (Hoboken) ; 294(1): 112-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21157922

ABSTRACT

Heme oxygenase-1 (HO-1), which catalyzes the degradation of heme to iron, carbon monoxide, and biliverdin, performs a cytoprotective function. Previous studies on the crystal structure of the human and rat HO-1 in complex with heme showed that Gly139His (G139H) and Gly143His (G143H) mutants have no HO activity. In the present study, we reported the effect of the G139H, G143H, and Ser142His mutants of mouse HO-1 on the HO reaction in vivo and in vitro. In vitro, of the mutant transfectants, only Ser142His catalyzed degradation of heme, retaining 31.7% of the wild-type mouse HO-1 activity, whereas G139H and G143H mutants exhibited no activity. In vivo, only Tg HO-1 G143H females presented with anemia, enlarged spleen and tissue iron overload, which was similar to HO-1(-/-) mice. The results suggested the critical role of Gly139 and Gly143 in maintaining HO-1 activity in vitro and the critical role of Gly143 in maintaining HO-1 activity in vivo.


Subject(s)
Amino Acid Substitution/genetics , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Anemia/enzymology , Anemia/genetics , Animals , Animals, Newborn , COS Cells , Chlorocebus aethiops , Female , Glycine/genetics , Heme Oxygenase-1/deficiency , Histidine/genetics , Iron Overload/enzymology , Iron Overload/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NIH 3T3 Cells , Serine/genetics , Splenomegaly/enzymology , Splenomegaly/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...