Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Cell Death Dis ; 12(3): 253, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33692332

ABSTRACT

Hepatocellular carcinoma (HCC) is a devastating malignancy without targeted therapeutic options. Our results indicated that the histone demethylase GASC1 signature is associated with later tumor stage and poorer survival in HCC patients. GASC1 depletion led to diminished HCC proliferation and tumor growth. A distinct heterogeneity in GASC1 levels was observed among HCC cell populations, predicting their inherent high or low tumor-initiating capacity. Mechanistically, GASC1 is involved in the regulation of several components of the Rho-GTPase signaling pathway including its downstream target ROCK2. GASC1 demethylase activity ensured the transcriptional repression of FBXO42, a ROCK2 protein-ubiquitin ligase, thereby inhibiting ROCK2 degradation via K63-linked poly-ubiquitination. Treatment with the GASC1 inhibitor SD70 impaired the growth of both HCC cell lines and xenografts in mice, sensitizing them to standard-of-care chemotherapy. This work identifies GASC1 as a malignant-cell-selective target in HCC, and GASC1-specific therapeutics represent promising candidates for new treatment options to control this malignancy.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Jumonji Domain-Containing Histone Demethylases/metabolism , Liver Neoplasms/enzymology , rho-Associated Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Enzyme Inhibitors/pharmacology , Enzyme Stability , F-Box Proteins/genetics , F-Box Proteins/metabolism , Gene Expression Regulation, Neoplastic , HEK293 Cells , Half-Life , Hep G2 Cells , Humans , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice, Inbred NOD , Mice, SCID , Proteolysis , Tumor Burden , Ubiquitination , Xenograft Model Antitumor Assays
2.
Adv Sci (Weinh) ; 7(21): 2001018, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33173725

ABSTRACT

The transcription factor SOX9 is frequently amplified in diverse advanced-stage human tumors. Its stability has been shown to be tightly controlled by ubiquitination-dependent proteasome degradation. However, the exact underlying molecular mechanisms remain unclear. This work reports that SOX9 protein abundance is regulated by the Cullin 3-based ubiquitin ligase KEAP1 via proteasome-mediated degradation. Loss-of-function mutations in KEAP1 compromise polyubiquitination-mediated SOX9 degradation, leading to increased protein levels, which facilitate tumorigenesis. Moreover, the loss of critical ubiquitination residues in SOX9, by either a SOX9 (ΔK2) truncation or K249R mutation, leads to elevated protein stability. Furthermore, it is shown that the KEAP1/SOX9 interaction is modulated by CKIγ-mediated phosphorylation. Importantly, it is demonstrated that DNA damage drugs, topoisomerase inhibitors, can trigger CKI activation to restore the KEAP1/SOX9 interaction and its consequent degradation. Collectively, herein the findings uncover a novel molecular mechanism through which SOX9 protein stability is negatively regulated by KEAP1 to control tumorigenesis. Thus, these results suggest that mitigating SOX9 resistance to KEAP1-mediated degradation can represent a novel therapeutic strategy for cancers with KEAP1 mutations.

3.
Article in English | MEDLINE | ID: mdl-32174890

ABSTRACT

Over the past decade, the increasing prevalence of obesity and its associated metabolic disorders constitutes one of the most concerning healthcare issues for countries worldwide. In an effort to curb the increased mortality and morbidity derived from the obesity epidemic, various therapeutic strategies have been developed by researchers. In the recent years, advances in the field of adipocyte biology have revealed that the thermogenic adipose tissue holds great potential in ameliorating metabolic disorders. Additionally, epigenetic research has shed light on the effects of histone acetylation on adipogenesis and thermogenesis, thereby establishing the essential roles which histone acetyltransferases (HATs) and histone deacetylases (HDACs) play in metabolism and systemic energy homeostasis. In regard to the therapeutic potential of thermogenic adipocytes for the treatment of metabolic diseases, herein, we describe the current state of knowledge of the regulation of thermogenic adipocyte differentiation and adaptive thermogenesis through histone acetylation. Furthermore, we highlight how different HATs and HDACs maintain the epigenetic transcriptional network to mediate the pathogenesis of various metabolic comorbidities. Finally, we provide insights into recent advances of the potential therapeutic applications and development of HAT and HDAC inhibitors to alleviate these pathological conditions.


Subject(s)
Adaptation, Physiological , Adipocytes/cytology , Adipogenesis , Cell Differentiation , Histones/chemistry , Thermogenesis , Acetylation , Adipocytes/physiology , Animals , Humans
4.
Gastroenterology ; 158(3): 664-678.e24, 2020 02.
Article in English | MEDLINE | ID: mdl-31678303

ABSTRACT

BACKGROUND & AIMS: Immune checkpoint inhibitors have some efficacy in the treatment of hepatocellular carcinoma (HCC). Programmed cell death 1 ligand 1 (PD-L1), expressed on some cancer cells, binds to the receptor programmed cell death 1 (PDCD1, also called PD1) on T cells to prevent their proliferation and reduce the antigen-tumor immune response. Immune cells that infiltrate some types of HCCs secrete interferon gamma (IFNG). Some HCC cells express myocyte enhancer factor 2D (MEF2D), which has been associated with shorter survival times of patients. We studied whether HCC cell expression of MEF2D regulates expression of PD-L1 in response to IFNG. METHODS: We analyzed immune cells from 20 fresh HCC tissues by flow cytometry. We analyzed 225 fixed HCC tissues (from 2 cohorts) from patients in China by immunohistochemistry and obtained survival data. We created mice with liver-specific knockout of MEF2D (MEF2DLPC-KO mice). We knocked out or knocked down MEF2D, E1A binding protein p300 (p300), or sirtuin 7 (SIRT7) in SMMC-7721, Huh7, H22, and Hepa1-6 HCC cell lines, some incubated with IFNG. We analyzed liver tissues from mice and cell lines by RNA sequencing, immunoblot, dual luciferase reporter, and chromatin precipitation assays. MEF2D protein acetylation and proteins that interact with MEF2D were identified by coimmunoprecipitation and pull-down assays. H22 cells, with MEF2D knockout or without (controls), were transplanted into BALB/c mice, and some mice were given antibodies to deplete T cells. Mice bearing orthotopic tumors grown from HCC cells, with or without knockout of SIRT7, were given injections of an antibody against PD1. Growth of tumors was measured, and tumors were analyzed by immunohistochemistry and flow cytometry. RESULTS: In human HCC specimens, we found an inverse correlation between level of MEF2D and numbers of CD4+ and CD8+ T cells; level of MEF2D correlated with percentages of PD1-positive or TIM3-positive CD8+ T cells. Knockout of MEF2D from H22 cells reduced their growth as allograft tumors in immune-competent mice but not in immune-deficient mice or mice with depletion of CD8+ T cells. When MEF2D-knockout cells were injected into immune-competent mice, they formed smaller tumors that had increased infiltration and activation of T cells compared with control HCC cells. In human and mouse HCC cells, MEF2D knockdown or knockout reduced expression of PD-L1. MEF2D bound the promoter region of the CD274 gene (encodes PD-L1) and activated its transcription. Overexpression of p300 in HCC cells, or knockout of SIRT7, promoted acetylation of MEF2D and increased its binding, along with acetylated histones, to the promoter region of CD274. Exposure of HCC cells to IFNG induced expression of p300 and its binding MEF2D, which reduced the interaction between MEF2D and SIRT7. MEF2D-induced expression of PD-L1 upon IFNG exposure was independent of interferon-regulatory factors 1 or 9. In HCC cells not exposed to IFNG, SIRT7 formed a complex with MEF2D that attenuated expression of PD-L1. Knockout of SIRT7 reduced proliferation of HCC cells and growth of tumors in immune-deficient mice. Compared with allograft tumors grown from control HCC cells, in immune-competent mice, tumors grown from SIRT7-knockout HCC cells expressed higher levels of PD-L1 and had reduced infiltration and activation of T cells. In immune-competent mice given antibodies to PD1, allograft tumors grew more slowly from SIRT7-knockout HCC cells than from control HCC cells. CONCLUSIONS: Expression of MEF2D by HCC cells increases their expression of PD-L1, which prevents CD8+ T-cell-mediated antitumor immunity. When HCC cells are exposed to IFNG, p300 acetylates MEF2D, causing it to bind the CD274 gene promoter and up-regulate PD-L1 expression. In addition to promoting HCC cell proliferation, SIRT7 reduced acetylation of MEF2D and expression of PD-L1 in HCC cells not exposed to IFNG. Strategies to manipulate this pathway might increase the efficacy of immune therapies for HCC.


Subject(s)
B7-H1 Antigen/genetics , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Sirtuins/genetics , Adolescent , Adult , Aged , Animals , CD4 Lymphocyte Count , CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/genetics , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/metabolism , Female , Gene Knockout Techniques , Hepatitis A Virus Cellular Receptor 2/metabolism , Humans , Immunocompetence , Interferon-gamma/pharmacology , Liver Neoplasms/pathology , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Male , Mice , Mice, Inbred BALB C , Middle Aged , Neoplasm Transplantation , Programmed Cell Death 1 Receptor/metabolism , Sirtuins/metabolism , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Young Adult
5.
Cancer Lett ; 463: 1-10, 2019 Oct 28.
Article in English | MEDLINE | ID: mdl-31394122

ABSTRACT

Hepatocellular carcinoma (HCC) is a highly malignant liver tumor. The presence of cancer stem cells (CSCs) figures prominently in tumor invasion, therapeutic resistance and tumor recurrence resulting in poor outcome and limited therapeutic options. Wnt/ß-catenin signaling is essential for cancer stem cell regulation and tumorigenesis in HCC, but its molecular mechanisms are not fully understood. Here, we demonstrate that ß-catenin is overexpressed in liver CSCs, and its expression level is positively correlated with SIRT1 in HCC specimens. SIRT1 regulates the protein stability of ß-catenin, thereby affecting the transcriptional activity of Wnt/ß-catenin signaling in liver CSCs. Mechanistically, we show that nuclear accumulation of ß-catenin results from deacetylation mediated by SIRT1. Further, nuclear ß-catenin promotes the transcription of Nanog to help maintain self-renewal of liver CSCs. Taken together, our findings indicate that the deacetylation of ß-catenin by SIRT1 represents a critical mechanism for regulating liver CSCs self-renewal and tumorigenesis. It provides an improved understanding of molecular mechanisms underlying ß-catenin activation and tumorigenesis in HCC.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Hepatocellular/metabolism , Gene Expression Regulation, Neoplastic/physiology , Liver Neoplasms/metabolism , Neoplastic Stem Cells/physiology , Sirtuin 1/physiology , beta Catenin/metabolism , Humans , Sirtuin 1/metabolism , Tumor Cells, Cultured
6.
Hepatology ; 70(4): 1197-1213, 2019 10.
Article in English | MEDLINE | ID: mdl-30901096

ABSTRACT

Metabolic reprogramming endows cancer cells with the ability to adjust metabolic pathways to support heterogeneously biological processes. However, it is not known how the reprogrammed activities are implemented during differentiation of cancer stem cells (CSCs). In this study, we demonstrated that liver CSCs relied on the enhanced mitochondrial function to maintain stemness properties, which is different from aerobic glycolysis playing main roles in the differentiated non-CSCs. We found that liver CSCs exhibit increased mitochondrial respiratory capacity and that complex-I of mitochondria was necessary for stemness properties of liver CSCs through regulation of mitochondrial respiration. Bioinformatics analysis reveals that mitochondrial ribosomal protein S5 (MRPS5) is closely related with the function of complex-I. Further experiments confirmed that MRPS5 promoted the production of nicotinamide adenine dinucleotide (NAD+ ), which is necessary for enhanced mitochondrial function in liver CSCs. MRPS5 played a critical role for liver CSCs to maintain stemness properties and to participate in tumor progression. Mechanistically, the acetylation status of MRPS5 is directly regulated by NAD+ dependent deacetylase sirtuin-1 (SIRT1), which is abundant in liver CSCs and decreased during differentiation. Deacetylated MRPS5 locates in mitochondria to promote the function complex-I and the generation of NAD+ to enhance mitochondrial respiration. Conversely, the acetylated MRPS5 gathered in nuclei leads to increased expression of glycolytic proteins and promotion of the Warburg Effect. Therefore, liver CSCs transform mitochondrial-dependent energy supply to a Warburg phenotype by the dual function of MRPS5. Clinical analysis of SIRT1 and MRPS5 expression in tumor tissues showed the SIRT1High /Cytoplasmic-MRPS5High profile was associated with patients with hepatocellular carcinoma with poor prognosis. Conclusion: SIRT1/MRPS5 axis participates in metabolic reprogramming to facilitate tumor progression and may serve as a promising therapeutic target of liver cancer.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cellular Reprogramming/genetics , Liver Neoplasms/genetics , Mitochondrial Proteins/genetics , NAD/metabolism , Ribosomal Proteins/genetics , Sirtuin 1/metabolism , Acetylation/drug effects , Animals , Carcinoma, Hepatocellular/pathology , Cell Differentiation/genetics , DNA Methylation/genetics , Humans , Liver Neoplasms/pathology , Mice , Mitochondria/metabolism , Neoplastic Stem Cells/metabolism , Sensitivity and Specificity , Tumor Cells, Cultured
7.
Sheng Wu Gong Cheng Xue Bao ; 34(5): 752-760, 2018 May 25.
Article in Chinese | MEDLINE | ID: mdl-29893083

ABSTRACT

This study was aimed to investigate the effect of Forkhead Box G1 (FOXG1) on the epithelial-mesenchymal transition (EMT) of colorectal cancer (CRC) cells and the underlying mechanism. For this purpose, FOXG1 lentiviral interference (shRNA) plasmid and expression plasmid were constructed. Western blotting was used to analyze the expression of FOXG1 protein in five CRC cells, namely RKO, SW480, SW620, LoVo and DLD-1. The shRNA fragment of FOXG1 (shFOXG1) was designed and synthesized. Recombinant plasmids were obtained with the aid of DNA recombination technique. Double digestion and sequencing were used to identify the recombinant plasmids, and then lentivirus packaging, purification and stable transfection were carried out. Additionally, stable CRC cell lines were screened out. The changes of FOXG1 knockdown and overexpression efficiency, E-cadherin, Vimentin, Fibronectin, Snail, Twist mRNA and protein were investigated respectively by Western blotting and qRT-PCR analysis. Furthermore, the changes of cell morphology after knockdown and cell migration ability were evaluated respectively with optical microscopy, scratch test and Transwell assay. FOXG1 had the highest protein expression in RKO and the lowest in DLD-1 among the five CRC cells. Compared with those of the control group, the cell morphology in FOXG1 knockdown RKO group was changed from spindle into round or polygonal shape, cell polarization was enhanced and tight junction assembly was acclerated while cell migration distance was noticeably decreased. Moreover, the number of cells invaded and migrated through chambers was significantly reduced. Among these key factors of EMT, the expression of E-cadherin was increased while the expressions of Vimentin, Fibronectin, Snail and Twist were decreased. The opposite was the case in the overexpressed FOXG1 group. The overexpression of FOXG1 in CRC promoted the invasion and metastasis of CRC cells and played a crucial role in regulating the EMT. Thus, FOXG1 might be a novel therapeutic target in CRC treatment.


Subject(s)
Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition , Forkhead Transcription Factors/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Nerve Tissue Proteins/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans
8.
Oncotarget ; 9(11): 9963-9974, 2018 Feb 09.
Article in English | MEDLINE | ID: mdl-29515783

ABSTRACT

Drug resistance is one of the main hurdles to overcome for the improvement of cancer patient survival. However, the underlying mechanisms remain largely unknown, and therapeutic options are limited. Here, we demonstrate a strong correlation between HMGA2 expression and chemosensitivity to 5-fluorouracil (5-FU), a widely used first-line systemic chemotherapy regimen for colorectal cancer (CRC) patients. Overexpression of HMGA2 enhances chemoresistance to 5-FU of CRC both in vitro and in vivo. Further experiments indicate that HMGA2 directly binds to the promoter of Dvl2 and induces its transcription, which leads to increased activation of the Wnt/ß-catenin pathway. Taken together, our data suggest that HMGA2 enhances the chemoresistance to 5-FU in CRC via activating the Dvl2/Wnt pathway. Therefore, HMGA2 may serve as a predictive biomarker and a potential therapeutic target in CRC.

9.
Cancer Res ; 78(4): 938-949, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29259010

ABSTRACT

Cancer stem-like cells (CSC) in hepatocellular carcinoma (HCC) are thought to mediate therapeutic resistance and poor survival outcomes, but their intrinsic and extrinsic control is not well understood. In this study, we found that the chromatin modification factor LSD1 is highly expressed in HCC CSC where it decreases during differentiation. LSD1 was responsible for maintaining CSC self-renewal and tumorigenicity in HCC, and its overexpression was sufficient to drive self-renewal of non-CSC. Levels of acetylated LSD1 were low in CSC with high LSD1 activity, and these CSC were capable of self-renewal. Notch signaling activated LSD1 through induction of the sirtuin SIRT1, leading to deacetylation and activation of LSD1 and CSC self-renewal. Notably, we found that LSD1 expression was increased in cancer-associated fibroblasts (CAF) as an upstream driver of Notch3-mediated CSC self-renewal. In clinical specimens of HCC, the presence of CAF, LSD1, and Notch3 strongly associated with poor patient survival. Overall, our results reveal that CAF-induced expression of Notch3 is responsible for LSD1 activation in CSC, driving their self-renewal in HCC.Significance: These seminal findings illuminate a complex pathway in the tissue microenvironment of liver cancer, which is responsible for orchestrating the self-renewal of stem-like cancer cells, with potential implications to improve therapy and limit relapses. Cancer Res; 78(4); 938-49. ©2017 AACR.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Carcinoma, Hepatocellular/metabolism , Histone Demethylases/metabolism , Liver Neoplasms/metabolism , Neoplastic Stem Cells/pathology , Receptor, Notch3/metabolism , Animals , Cancer-Associated Fibroblasts/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/physiology , Heterografts , Histone Demethylases/biosynthesis , Histone Demethylases/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Receptor, Notch3/genetics , Signal Transduction
10.
Mol Med Rep ; 16(2): 2172-2178, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28656300

ABSTRACT

The medicinal fungus Paecilomyces tenuipes exhibits a variety of pharmacological effects, including antidepressive effects. The chronic unpredictable mild stress (CUMS)­induced rat model has served an important role in studies involving antidepressants screening. The aim of the present study was to evaluate the antidepressant­like activity of P. tenuipes N45 aqueous extract (PTNE) in a CUMS­induced rat model of behavioral despair depression. Following 4 weeks of PTNE treatment, behavioral tests were conducted to investigate the antidepressant­like activities, and the levels of neurotransmitters and hormones in blood and hypothalamus were measured. The results demonstrated that PTNE treatment significantly increased movement in the forced running test, whereas the immobility time was reduced in the hotplate test and the forced swim test in depression­model rats. PTNE treatment was able to normalize the levels of hormones and neurotransmitters in serum and hypothalamus of CUMS rats. The data demonstrated that PTNE treatment may be a potential pharmaceutical agent in treatment­resistant depression, and the effects of PTNE may be partly mediated through normalizing the levels of neurotransmitters.


Subject(s)
Depressive Disorder/prevention & control , Paecilomyces/chemistry , Stress, Psychological , Adrenocorticotropic Hormone/analysis , Adrenocorticotropic Hormone/blood , Animals , Behavior, Animal/drug effects , Depressive Disorder/etiology , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Enzyme-Linked Immunosorbent Assay , Hypothalamus/metabolism , Male , Maze Learning/drug effects , Neurotransmitter Agents/analysis , Neurotransmitter Agents/blood , Neurotransmitter Agents/metabolism , Paecilomyces/metabolism , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Glucocorticoid/analysis , Receptors, Glucocorticoid/blood
11.
Cancer Lett ; 400: 117-126, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28478181

ABSTRACT

Myocyte enhancer factor 2D (MEF2D) is involved in many aspects of cancer progression, including cell proliferation, invasion, and migration. However, little is known about the role of MEF2D in tumor angiogenesis. Using clinical specimens, colorectal cancer (CRC) cell lines and a mouse model in the present study, we found that MEF2D expression was positively correlated with CD31-positive microvascular density in CRC tissues. MEF2D promoted tumor angiogenesis in vitro and in vivo and induced the expression of proangiogenic cytokines in CRC cells. MEF2D was found to be a downstream effector of hypoxia-inducible factor (HIF)-1α in the induction of tumor angiogenesis. HIF-1α transactivates MEF2D expression by binding to the MEF2D gene promoter. These results demonstrate that the HIF-1α/MEF2D axis can serve as a therapeutic target for the treatment of CRC.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neovascularization, Pathologic , Animals , Binding Sites , Biomarkers, Tumor/genetics , Caco-2 Cells , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Culture Media, Conditioned/metabolism , Cytokines/metabolism , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Microvessels/metabolism , Microvessels/pathology , Middle Aged , Paracrine Communication , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Prognosis , Promoter Regions, Genetic , RNA Interference , Signal Transduction , Time Factors , Transcriptional Activation , Transfection , Tumor Burden , Tumor Hypoxia , Tumor Microenvironment
12.
Mol Med Rep ; 15(5): 2807-2813, 2017 May.
Article in English | MEDLINE | ID: mdl-28447736

ABSTRACT

Due to its pharmacological activities, Paecilomyces tenuipes has previously been used as a folk medicine in Asia. The primary aim of the present study was to investigate the hypoglycemic, hypolipidemic and anti­nephritic effects of P. tenuipes N45 aqueous extracts (PTNE) in a high fat diet/streptozotocin­induced diabetic rat model. The rats were treated with 120 mg/kg of metformin or 0.04, 0.2 or 1.0 g/kg PTNE for 4 weeks. The hypoglycemic activity of PTNE was confirmed by the observation of reduced fasting blood glucose level and by partially normalized oral glucose tolerance. PTNE reduced total cholesterol and triglyceride content, and balanced the levels of low­density and high­density lipoproteins. The suppressive effects of PTNE on creatinine, blood urea nitrogen, interleukin (IL)­2, IL­6 and nuclear factor­κB levels indicated its ability to provide protection against diabetic nephropathy. PTNE treatment increased superoxide dismutase, malondialdehyde and glutathione peroxidase levels, suggesting that its anti­diabetic and anti­nephropathic activities may be associated with the prevention of oxidative damage during type 2 diabetic mellitus. The findings of the present study provided experimental evidence for the application of Paecilomyces tenuipes N45 on the treatment of type 2 diabetic mellitus.


Subject(s)
Ascomycota/chemistry , Complex Mixtures/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Hypoglycemic Agents/pharmacology , Hypolipidemic Agents/pharmacology , Animals , Complex Mixtures/chemistry , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Hypoglycemic Agents/chemistry , Hypolipidemic Agents/chemistry , Interleukin-2/metabolism , Interleukin-6/metabolism , NF-kappa B/metabolism , Rats
13.
Oncotarget ; 8(21): 34362-34373, 2017 May 23.
Article in English | MEDLINE | ID: mdl-28416766

ABSTRACT

Abnormal activation of Notch signaling is involved in the etiology of various diseases, including cancer, but the association between Notch3 expression in urothelial cancer and clinical outcome remains unclear, and the molecular mechanisms underlying Notch3 signaling activation are not well defined. In this study we examined 59 urothelial cancer patients and found that Notch3 was more highly expressed in human urothelial cancer tissues than in non-tumorous bladder tissue samples, with Notch3 overexpression being associated with poor clinical outcome. Notch3 knockdown resulted in decreased proliferation of urothelial cancer cells in vitro and decreased xenograft tumor growth in vivo. In addition, Notch3 knockdown rendered urothelial cancer cells more sensitive to cisplatin. Furthermore, suberoylanilide hydroxamic acid (SAHA, a histone deacetylase [HDAC] inhibitor) induced acetylation of NOTCH3, downregulated Notch 3, prevented urothelial cancer cell proliferation, and induced cell cycle arrest. Taken together, these data suggested that Notch 3 overexpression promotes growth and chemoresistance in urothelial cancer.


Subject(s)
Carcinoma, Transitional Cell/pathology , Drug Resistance, Neoplasm , Receptor, Notch3/metabolism , Up-Regulation , Urinary Bladder Neoplasms/pathology , Animals , Carcinoma, Transitional Cell/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/administration & dosage , Cisplatin/pharmacology , Disease Progression , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Neoplasm Transplantation , Prognosis , Survival Analysis , Urinary Bladder Neoplasms/metabolism
14.
Exp Ther Med ; 13(1): 352-358, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28123514

ABSTRACT

Due to substantial morbidity and complications including nephropathy, a search for alternative treatment of diabetes mellitus is urgently required. The present study aimed to investigate the hypoglycemic and anti-diabetic nephropathy activities of polysaccharides separated from Auricularia auricular (AAP). Diet streptozotocin (STZ)-induced diabetic Sprague-Dawley rats were orally treated with metformin (100 mg/kg; positive control) and AAP (100 and 400 mg/kg) for four weeks, and parameters in the serum and liver associated with blood glucose, free radicals and nephropathy were determined. Similar to metformin, AAP treatment strongly reduced blood glucose levels by promoting glucose metabolism. The anti-oxidative activity of AAP, which was indicated by the modulation of superoxide dismutase, glutathione peroxidase, reactive oxygen species and methane dicarboxylic aldehyde levels in serum, was observed in diabetic rats. Furthermore, the regulatory effects of AAP on blood urea nitrogen, creatinine, uric protein and inflammatory-related factors revealed its protection against diabetic nephropathy. The present data suggests that AAP-mediated anti-diabetic and anti-nephritic effects are partially associated with their modulations on the anti-oxidative system and nuclear factor kappa B-related signaling pathway. In conclusion, AAP has potential to be a novel source of treatments for diabetes.

15.
Oncotarget ; 8(66): 110187-110200, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29299140

ABSTRACT

Cancer stem cells (CSCs) are thought to be responsible for tumor invasion, metastasis, and recurrence. We previously showed that the pluripotency factor Nanog not only serves as a novel biomarker of CSCs but also potentially plays a crucial role in maintaining the self-renewal ability of liver CSCs. However, how CSCs maintain Nanog gene expression has not been elucidated. Here, we demonstrated that microRNA-449a (miR-449a) is overexpressed in poorly differentiated hepatocellular carcinoma tissues, drug-resistant liver cancer cells, cultured liver tumorspheres, and Nanog-positive liver cancer cells. The upregulation of miR-449a in non-CSCs increased stemness, whereas the downregulation of miR-449a in Nanog-positive CSCs reduced stemness. Furthermore, transcription factor 3 (TCF3), a target of miR-449a, could downregulate Nanog expression, and restoring TCF3 expression in miR-449a-expressing Nanog-negative cells abrogated cellular stemness. These data establish that the miR449a-TCF3-Nanog axis maintains stemness in liver CSCs.

16.
Cancer Res ; 76(17): 5054-67, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27364559

ABSTRACT

Epithelial-mesenchymal transition (EMT) is an essential mechanism of metastasis, including in colorectal cancer. Although EMT processes are often triggered in cancer cells by their surrounding microenvironment, how EMT-relevant genes control these processes is not well understood. In multiple types of cancers, the transcription factor MEF2D has been implicated in cell proliferation, but its contributions to metastasis have not been addressed. Here, we show MEF2D is overexpressed in clinical colorectal cancer tissues where its high expression correlates with metastatic process. Functional investigations showed that MEF2D promoted cancer cell invasion and EMT and that it was essential for certain microenvironment signals to induce EMT and metastasis in vivo Mechanistically, MEF2D directly regulated transcription of the EMT driver gene ZEB1 and facilitated histone acetylation at the ZEB1 promoter. More importantly, MEF2D responded to various tumor microenvironment signals and acted as a central integrator transducing multiple signals to activate ZEB1 transcription. Overall, our results define a critical function for MEF2D in upregulating EMT and the metastatic capacity of colorectal cancer cells. Further, they offer new insights into how microenvironment signals activate EMT-relevant genes and deepen the pathophysiologic significance of MEF2D, with potential implications for the prevention and treatment of metastatic colorectal cancer. Cancer Res; 76(17); 5054-67. ©2016 AACR.


Subject(s)
Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/physiology , Tumor Microenvironment/physiology , Zinc Finger E-box-Binding Homeobox 1/metabolism , Adult , Aged , Animals , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Female , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Immunohistochemistry , Kaplan-Meier Estimate , MEF2 Transcription Factors/metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Metastasis/pathology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology
17.
Hepatology ; 64(3): 814-27, 2016 09.
Article in English | MEDLINE | ID: mdl-27312708

ABSTRACT

UNLABELLED: Hepatocellular carcinoma (HCC) is a highly aggressive liver tumor containing cancer stem cells (CSCs), which participate in tumor invasion, therapeutic resistance, and tumor relapse leading to poor outcome and limited therapeutic options. Histone deacetylatase sirtuin 1 (SIRT1) has been shown to be up-regulated in human cancers; however, its role in liver CSCs is unknown. In this study, we explored the biological functions of SIRT1 in liver CSCs. Our data show that SIRT1 is highly expressed in liver CSCs and decreases during differentiation. In addition, high levels of SIRT1 predict a decreased probability of survival in patients with HCC. SIRT1 is responsible for the maintenance of self-renewal and tumorigenicity of liver CSCs, and overexpression of exogenous SIRT1 can restore self-renewal of non-CSCs. We demonstrated that SOX2 is a main downstream regulator of SIRT1-mediated self-renewal and tumorigenicity potential of liver CSCs. Mechanistically, SIRT1 regulates transcription of the SOX2 gene by way of chromatin-based epigenetic changes, which are dependent on DNA methylation. This effect is achieved by alternation of histone modification and interaction with DNA methyltransferase 3A, resulting in hypermethylation of SOX2 promoter. Furthermore, we demonstrated that insulin growth factor signaling plays an important role in maintaining SIRT1 expression through increased SIRT1 protein stability. CONCLUSIONS: These findings highlight the importance of SIRT1 in the biology of liver CSCs and suggest that SIRT1 may serve as a molecular target for HCC therapy. (Hepatology 2016;64:814-827).


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Self Renewal , Liver Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , SOXB1 Transcription Factors/metabolism , Sirtuin 1/metabolism , Animals , DNA Methyltransferase 3A , Epigenesis, Genetic , Female , HEK293 Cells , Humans , Male , Mice, Inbred NOD , Mice, SCID , Middle Aged , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , Somatomedins/metabolism
18.
Oxid Med Cell Longev ; 2016: 9685257, 2016.
Article in English | MEDLINE | ID: mdl-27274781

ABSTRACT

Cordyceps militaris has long been used as a crude drug and folk tonic food in East Asia. The present study aims to evaluate the antidiabetic and antinephritic effects of the aqueous extract of the Cordyceps militaris fruit body (CM) in diet-streptozotocin- (STZ-) induced diabetic rats. During four weeks of continuous oral administration of CM at doses of 0.5, 1.0, and 2.0 g/kg and metformin at 100 mg/kg, the fasting blood glucose and bodyweight of each rat were monitored. Hypoglycemic effects of CM on diabetic rats were indicated by decreases in plasma glucose, food and water intake, and urine output. The hypolipidemic activity of CM was confirmed by the normalization of total cholesterol, triglycerides, and low- and high-density lipoprotein cholesterol in diabetic rats. Inhibitory effects on albuminuria, creatinine, urea nitrogen, and n-acetyl-ß-d-glucosaminidase verified CM's renal protective activity in diabetic rats. Furthermore, CM exerted beneficial modulation of inflammatory factors and oxidative enzymes. Compared with untreated diabetic rats, CM decreased the expression of phosphor-AKT and phosphor-GSK-3ß in the kidneys. Altogether, via attenuating oxidative stress, CM displayed antidiabetic and antinephritic activities in diet-STZ-induced diabetic rats.


Subject(s)
Cordyceps/chemistry , Diabetes Mellitus, Experimental/drug therapy , Hypoglycemic Agents/therapeutic use , Plant Extracts/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Blood Glucose/analysis , Blood Urea Nitrogen , Body Weight/drug effects , Cordyceps/metabolism , Cytokines/analysis , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/pathology , Enzyme-Linked Immunosorbent Assay , Fruit/chemistry , Fruit/metabolism , Glucose Tolerance Test , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Kidney/metabolism , Kidney/pathology , Lipids/blood , Male , Plant Extracts/chemistry , Plant Extracts/pharmacology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Streptozocin/toxicity
19.
Food Funct ; 7(4): 2006-15, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27008597

ABSTRACT

Membranous glomerulonephritis (MGN) is a common pathogenesis of nephritic syndrome in adult patients. Nuclear factor kappa B (NF-κB) serves as the main transcription factor for the inflammatory response mediated nephropathy. Cordyceps militaris, containing various pharmacological components, has been used as a kind of crude drug and folk tonic food for improving immunity and reducing inflammation. The current study aims to investigate the renoprotective activity of Cordyceps militaris aqueous extract (CM) in the cationic bovine serum albumin (C-BSA)-induced rat model of membranous glomerulonephritis. Significant renal dysfunction was observed in MGN rats; comparatively, 4-week CM administration strongly decreased the levels of 24 h urine protein, total cholesterol, triglyceride, blood urea nitrogen and serum creatinine, and increased the levels of serum albumin and total serum protein. Strikingly, recovery of the kidney histological architecture was noted in CM-treated MGN rats. A significant improvement in the glutathione peroxidase and superoxide dismutase levels, and a reduced malondialdehyde concentration were observed in the serum and kidney of CM-treated rats. Altered levels of inflammatory cytokines including interleukins, monocyte chemoattractant protein-1, intercellular adhesion molecule 1, vascular adhesion molecule 1, tumor necrosis factor-α, 6-keto-prostaglandin F1α, and nuclear transcriptional factor subunit NF-κB p65 reverted to normal levels upon treatment with CM. The present data suggest that CM protects rats against membranous glomerulonephritis via the normalization of NF-κB activity, thereby inhibiting oxidative damage and reducing inflammatory cytokine levels, which further provide experimental evidence in support of the clinical use of CM as an effective renoprotective agent.


Subject(s)
Biological Factors/administration & dosage , Cordyceps/chemistry , Fruiting Bodies, Fungal/chemistry , Glomerulonephritis, Membranous/drug therapy , Kidney/immunology , NF-kappa B/metabolism , Oxidative Stress/drug effects , Animals , Glomerulonephritis, Membranous/immunology , Glomerulonephritis, Membranous/metabolism , Humans , Kidney/drug effects , Male , NF-kappa B/genetics , Rats , Rats, Wistar , Signal Transduction/drug effects , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/metabolism
20.
Oncotarget ; 7(15): 20597-611, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26967560

ABSTRACT

Hepatocellular carcinoma (HCC) is the third leading cause of cancer death. This high mortality has been commonly attributed to the presence of residual cancer stem cells (CSCs). Meanwhile, MEK1 signaling is regarded as a key molecular in HCC maintenance and development. However, nobody has figured out the particular mechanisms that how MEK1 signaling regulates liver CSCs self-renewal. In this study, we show that inhibition or depletion of MEK1 can significantly decrease liver CSCs self-renewal and tumor growth both in vitro and vivo conditions. Furthermore, we demonstrate that MEK1 signaling promotes liver CSCs self-renewal and tumorigenicity by maintaining SIRT1 level. Mechanistically, MEK1 signaling keeps SIRT1 protein stabilization through activating SIRT1 ubiquitination, which inhibits proteasomal degradation. Clinical analysis shows that patients co-expression of MEK1 and SIRT1 are associated with poor survival. Our finding indicates that MEK1-SIRT1 can act as a novel diagnostic biomarker and inhibition of MEK1 may be a viable therapeutic option for targeting liver CSCs treatment.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Self Renewal , Liver Neoplasms/pathology , MAP Kinase Kinase 1/metabolism , Neoplastic Stem Cells/pathology , Sirtuin 1/chemistry , Animals , Apoptosis , Biomarkers, Tumor , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , Prognosis , Protein Stability , Signal Transduction , Sirtuin 1/metabolism , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...