Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
J Biochem Mol Toxicol ; 38(2): e23645, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38348716

ABSTRACT

Prostate cancer (PCa) is an extremely common genitourinary malignancy among elderly men. Many evidence have shown the efficacy of curcumin (CUR) in inhibiting the progression of PCa. However, the pharmacological function of CUR in PCa is still not quite clear. In this research, CUR was found to suppress the proliferation and enhance the apoptotic rate in in vitro PCa cell models in a dose- and time-dependent manner. In a xenograft animal model, the administration of CUR contributed to a significant decrease in the growth of the xenograft tumor induced by the transplanted PC-3 cells. Ubiquitin-conjugating enzyme E2 C is implicated in the modulation of multiple types of cancers. In humans, the expression levels of UBE2C are significantly higher in PCa versus benign prostatic hyperplasia. Treatment with CUR decreased the expression of UBE2C, whereas it increased miR-483-3p expression. In contrast with the control mice, the CUR-treated mice showed a significant reduction in UBE2C and Ki-67 in PCa cells. The capability of proliferation, migration, and invasion of PCa cells was inhibited by the knockdown of UBE2C mediated by siRNA. Furthermore, dual luciferase reporter gene assay indicated the binding of miR-483-3p to UBE2C. In summary, CUR exerts its antitumor effects through regulation of the miR-483-3p/UBE2C axis by decreasing UBE2C and increasing miR-483-3p. The findings may also provide new molecular markers for PCa diagnosis and treatment.


Subject(s)
Curcumin , MicroRNAs , Prostatic Neoplasms , Male , Humans , Animals , Mice , Aged , MicroRNAs/genetics , MicroRNAs/metabolism , Curcumin/pharmacology , Curcumin/therapeutic use , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Cell Line, Tumor , Apoptosis/genetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Disease Models, Animal , Cell Proliferation/genetics , Cell Movement/genetics , Gene Expression Regulation, Neoplastic
2.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 52(5): 594-604, 2023 Sep 28.
Article in English, Chinese | MEDLINE | ID: mdl-37916309

ABSTRACT

OBJECTIVES: To explore the mechanism of transforming growth factor-ß1 (TGF-ß1) induce renal fibrosis. METHODS: Renal fibroblast NRK-49F cells treated with and without TGF-ß1 were subjected to RNA-seq analysis. DESeq2 was used for analysis. Differentially expressed genes were screened with the criteria of false discovery rate<0.05 and l o g 2 F C >1. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed for differentially expressed genes. Genes encoding transcription factors were further screened for differential expression genes. Then, the expression of these genes during renal fibrosis was verified using unilateral ureteral obstruction (UUO)-induced mouse renal fibrosis model and a public gene expression dataset (GSE104954). RESULTS: After TGF-ß1 treatment for 6, 12 and 24 h, 552, 1209 and 1028 differentially expressed genes were identified, respectively. GO analysis indicated that these genes were significantly enriched in development, cell death, and cell migration. KEGG pathway analysis showed that in the early stage of TGF-ß1 induction (TGF-ß1 treatment for 6 h), the changes in Hippo, TGF-ß and Wnt signaling pathways were observed, while in the late stage of TGF-ß1 induction (TGF-ß1 treatment for 24 h), the changes of extracellular matrix-receptor interaction, focal adhesion and adherens junction were mainly enriched. Among the 291 up-regulated differentially expressed genes treated with TGF-ß1 for 6 h, 13 genes (Snai1, Irf8, Bhlhe40, Junb, Arid5a, Vdr, Lef1, Ahr, Foxo1, Myc, Tcf7, Foxc2, Glis1) encoded transcription factors. Validation in a cell model showed that TGF-ß1 induced expression of 9 transcription factors (encoded by Snai1, Irf8, Bhlhe40, Junb, Arid5a, Vdr, Lef1, Myc, Tcf7), while the expression levels of the other 4 genes did not significantly change after TGF-ß1 treatment. Validation results in UUO-induced mouse renal fibrosis model showed that Snai1, Irf8, Bhlhe40, Junb, Arid5a, Myc and Tcf7 were up-regulated after UUO, Vdr was down-regulated and there was no significant change in Lef1. Validation based on the GSE104954 dataset showed that IRF8 was significantly overexpressed in the renal tubulointerstitium of patients with diabetic nephropathy or IgA nephropathy, MYC was highly expressed in diabetic nephropathy, and the expressions of the other 7 genes were not significantly different compared with the control group. CONCLUSIONS: TGF-ß1 induces differentially expressed genes in renal fibroblasts, among which Irf8 and Myc were identified as potential targets of chronic kidney disease and renal fibrosis.


Subject(s)
Diabetic Nephropathies , Ureteral Obstruction , Mice , Animals , Humans , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factor beta1/metabolism , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Transcriptome , Signal Transduction , Kidney , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology , Fibrosis , Interferon Regulatory Factors , Transforming Growth Factor beta/metabolism , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism
3.
J Clin Transl Res ; 9(3): 182-194, 2023 Jun 29.
Article in English | MEDLINE | ID: mdl-37275579

ABSTRACT

Background and Aim: Hepatocellular carcinoma (HCC) is one of the ten most common malignant tumors in the world, and it is a major problem in the world. Traditional Chinese medicine (TCM) has many advantages in the prevention and treatment of HCC, but its complicated mechanism of action is difficult to clarify, which limits its research and development. The continuous development of bioinformation technology provides new methods and opportunities for the research of TCM. This study used modern network pharmacology and bioinformatic methods to explore the possible molecular mechanism of the Chinese herbal compound Fuzheng Xiaoliu Granule (FZXLG) to treat HCC, to provide a theoretical basis for their clinical application and basic research, to promote the modernization of TCM, and to promote its worldwide application. Methods: The active ingredients of FZXLG were collected and screened through TCMSP, BATMAN-TCM, and other databases. The targets of FZXLG were predicted by PubChem and SwissTargetPrediction; HCC disease-related targets were obtained by GeneCards, OMIM, and other disease databases, and the potential gene targets of FZXLG for HCC treatment were screened. The "Prescription-TCMs-Ingredients-Targets" network of FZXLG for the treatment of HCC was constructed, along with the screening of core effective components. The differentially expressed genes (DEGs) of HCC tumor and non-tumor adjacent tissues combined with clinical data in the TCGA database were analyzed to obtain the prognostic genes of HCC. Then, FZXLG genes affecting HCC prognosis were screened and further screening the core target genes. The correlation between core gene expression with prognosis, immune cell infiltration, and immunohistochemical changes in HCC patients was studied. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis and Gene Ontology enrichment analysis of the FZXLG genes affecting HCC prognosis were performed using DAVID database. AutoDockTools software was then used for molecular docking verification. Results: The ten core effective ingredients of FZXLG for HCC treatment included multiple flavonoids ingredients such as quercetin, luteolin, and formononetin. 11 core targets of FZXLG affecting the prognosis of HCC were screened, among which estrogen receptor 1 (ESR1) and catalase (CAT) were favorable prognostic factors, while EGF, MMP9, CCNA2, CCNB1, CDK1, CHEK1, and E2F1 were adverse prognostic factors. MMP9 and EGF were positively correlated with six TIIC subsets. The different expression levels of CAT, PLG, AR, MMP9, CCNA2, CCNB1, CDK1, and E2F1 were correlated with the immunohistochemical staining changes in normal liver and liver cancer. KEGG pathway enrichment analysis yielded 33 pathways including cell cycle, p53, hepatitis B, and other signaling pathways. Molecular docking verified that the main core components had good binding to the protective prognostic core targets ESR1 and CAT. Conclusions: FZXLG may treat HCC through multiple ingredients, multiple targets, and multiple pathways, affecting the prognosis, immune microenvironment, and immunohistochemical changes of HCC. Relevance for Patients: FZXLG is a Chinese herbal compound for the treatment of HCC, with significant clinical efficacy. However, the mechanism of action is unclear and lacks theoretical support, which limits its popularization application. This study preliminarily revealed its molecular mechanism, providing a theoretical basis for its clinical application, which can better guide its clinical popularization application, and also provide a new strategy for the treatment of HCC.

4.
Cancer Prev Res (Phila) ; 16(6): 321-332, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36867722

ABSTRACT

Suberoylanilide hydroxamic acid (SAHA) is a histone deacetylase (HDAC) inhibitor with anticancer effects via epigenetic and non-epigenetic mechanisms. The role of SAHA in metabolic rewiring and epigenomic reprogramming to inhibit pro-tumorigenic cascades in lung cancer remains unknown. In this study, we aimed to investigate the regulation of mitochondrial metabolism, DNA methylome reprogramming, and transcriptomic gene expression by SAHA in lipopolysaccharide (LPS)-induced inflammatory model of lung epithelial BEAS-2B cells. LC/MS was used for metabolomic analysis, while next-generation sequencing was done to study epigenetic changes. The metabolomic study reveals that SAHA treatment significantly regulated methionine, glutathione, and nicotinamide metabolism with alteration of the metabolite levels of methionine, S-adenosylmethionine, S-adenosylhomocysteine, glutathione, nicotinamide, 1-methylnicotinamide, and nicotinamide adenine dinucleotide in BEAS-2B cells. Epigenomic CpG methyl-seq shows SAHA revoked a list of differentially methylated regions in the promoter region of the genes, such as HDAC11, miR4509-1, and miR3191. Transcriptomic RNA sequencing (RNA-seq) reveals SAHA abrogated LPS-induced differentially expressed genes encoding proinflammatory cytokines, including interleukin 1α (IL1α), IL1ß, IL2, IL6, IL24, and IL32. Integrative analysis of DNA methylome-RNA transcriptome displays a list of genes, of which CpG methylation correlated with changes in gene expression. qPCR validation of transcriptomic RNA-seq data shows that SAHA treatment significantly reduced the LPS-induced mRNA levels of IL1ß, IL6, DNA methyltransferase 1 (DNMT1), and DNMT3A in BEAS-2B cells. Altogether, SAHA treatment alters the mitochondrial metabolism, epigenetic CpG methylation, and transcriptomic gene expression to inhibit LPS-induced inflammatory responses in lung epithelial cells, which may provide novel molecular targets to inhibit the inflammation component of lung carcinogenesis. PREVENTION RELEVANCE: Inflammation increases the risk of lung cancer and blocking inflammation could reduce the incidence of lung cancer. Herein, we demonstrate that histone deacetylase inhibitor suberoylanilide hydroxamic acid regulates metabolic rewiring and epigenetic reprogramming to attenuate lipopolysaccharide-driven inflammation in lung epithelial cells.


Subject(s)
Lipopolysaccharides , Lung Neoplasms , Humans , Vorinostat , Lipopolysaccharides/pharmacology , Interleukin-6 , Transcriptome , Hydroxamic Acids/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Lung , Inflammation , DNA , Epithelial Cells , Glutathione/genetics , Methionine
5.
Chem Res Toxicol ; 35(7): 1220-1233, 2022 07 18.
Article in English | MEDLINE | ID: mdl-35700067

ABSTRACT

Sulforaphane (SFN) is a potent anticancer agent which could protect the skin from ultraviolet (UV) radiation-induced insults. Currently, the metabolic rewiring and epigenetic reprograming induced by UVB and the role of SFN in UVB-mediated skin cell transformation remain largely unknown. Herein, we study the metabolome, epigenome, and transcriptome of human keratinocytes (HaCaT cells) exposed to UVB with or without SFN using liquid chromatography-mass spectroscopy, DNA methylation sequencing, and RNA sequencing. UVB increases intracellular reactive oxygen species (ROS) and SFN enhances ROS acutely in post-UVB-exposed HaCaT cells. UVB and SFN alter multiple metabolites and metabolism-related signaling pathways. Pathway analysis shows that UVB impacts numerous signaling pathways including STAT3, inhibition of matrix metalloproteases, and TGF-ß, among others. DNA/CpG methylation analysis shows that SFN could partially reverse some of the alterations of UVB-induced CpG methylome. Integrating RNA-seq and Methyl-seq data, starburst plots show the correlation of mRNA expression and CpG methylation status. The potential linkages between the metabolome, CpG methylome, and transcriptome suggest that metabolites produced during metabolism act as cofactors or substrates for catalytic epigenetic modification and transcriptional regulation. These results indicate that UVB drives metabolic rewiring, epigenetic reprograming, and phenotypic transcriptomic alterations and SFN would block or attenuate many of these aberrations, potentially contributing to the overall protective effect of SFN against UVB-induced skin damage.


Subject(s)
Isothiocyanates , Keratinocytes , Apoptosis , Epigenesis, Genetic , Humans , Isothiocyanates/metabolism , Isothiocyanates/pharmacology , Reactive Oxygen Species/metabolism , Sulfoxides , Ultraviolet Rays
6.
AAPS J ; 24(1): 30, 2022 01 18.
Article in English | MEDLINE | ID: mdl-35043283

ABSTRACT

Fucoxanthin (FX) is a carotenoid with many pharmaceutical properties due to its antioxidant/anti-inflammatory and epigenetic effects. NFE2L2 is involved in the defense against oxidative stress/inflammation-mediated diseases, like anticancer effects elicited by phytochemicals including FX. However, the role of FX and NFE2L2 in metabolic rewiring, epigenomic reprogramming, and transcriptomic network in blocking pro-tumorigenic signaling and eliciting cancer-protective effects remains unknown. Herein, we utilized multi-omics approaches to evaluate the role of NFE2L2 and the impact of FX on tumor promoter TPA-induced skin cell transformation. FX blocked TPA-induced ROS and oxidized GSSG/reduced GSH in Nfe2l2wild-type(WT) but not Nfe2l2-knockdown (KD) cells. Both Nfe2l2 KD and TPA altered cellular metabolisms and metabolites which are tightly coupled to epigenetic machinery. The suppressive effects of FX on TPA-enhancedSAM/SAH was abrogated by Nfe2l2 KD indicating Nfe2l2 plays a critical role in FX-mediated metabolic rewiring and its potential consequences on epigenetic reprogramming. Epigenomic CpG methyl-seq revealed that FX attenuated TPA-induced differentially methylated regions (DMRs) of Uhrf1 and Dnmt1 genes. Transcriptomic RNA-seq showed that FX abrogated TPA-induced differentially expressed genes (DEGs) of Nfe2l2-related genes Nqo1, Ho1, and Keap1. Associative analysis of DEGs and DMRs identified that the mRNA expressions of Uhrf1 and Dnmt1 were correlated with the promoter CpG methylation status. Chromatin immunoprecipitation assay showed that FX restored Uhrf1 expression by regulating H3K27Me3 enrichment in the promoter region. In this context, FX/Nfe2l2's redox signaling drives metabolic rewiring causing epigenetic and transcriptomic reprogramming potentially contributing to the protection of TPA-induced JB6 cellular transformation skin cancer model. Graphical abstract.


Subject(s)
Epigenesis, Genetic , NF-E2-Related Factor 2/genetics , Skin Neoplasms/prevention & control , Xanthophylls/pharmacology , Animals , Antioxidants/pharmacology , Cell Line , Cell Transformation, Neoplastic/drug effects , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Mice , Oxidation-Reduction/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate
7.
Free Radic Biol Med ; 179: 328-336, 2022 02 01.
Article in English | MEDLINE | ID: mdl-33359432

ABSTRACT

Biological redox signaling plays an important role in many diseases. Redox signaling involves reductive and oxidative mechanisms. Oxidative stress occurs when reductive mechanism underwhelms oxidative challenges. Cellular oxidative stress occurs when reactive oxygen/nitrogen species (RO/NS) exceed the cellular reductive/antioxidant capacity. Endogenously produced RO/NS from mitochondrial metabolic citric-acid-cycle coupled with electron-transport-chain or exogenous stimuli trigger cellular signaling events leading to homeostatic response or pathological damage. Recent evidence suggests that RO/NS also modulate epigenetic machinery driving gene expression. RO/NS affect DNA methylation/demethylation, histone acetylation/deacetylation or histone methylation/demethylation. Many health beneficial phytochemicals possess redox capability that counteract RO/NS either by directly scavenging the radicals or via inductive mechanism of cellular defense antioxidant/reductive enzymes. Amazingly, these phytochemicals also possess epigenetic modifying ability. This review summarizes the latest advances on the interactions between redox signaling, mitochondrial metabolism, epigenetics and redox active phytochemicals and the future challenges of integrating these events in human health.


Subject(s)
Epigenesis, Genetic , Signal Transduction , Humans , Oxidation-Reduction , Oxidative Stress , Phytochemicals/pharmacology
8.
Mol Carcinog ; 61(1): 111-121, 2022 01.
Article in English | MEDLINE | ID: mdl-34727410

ABSTRACT

Ursolic acid (UA) is a triterpenoid phytochemical with a strong anticancer effect. The metabolic rewiring, epigenetic reprogramming, and chemopreventive effect of UA in prostate cancer (PCa) remain unknown. Herein, we investigated the efficacy of UA in PCa xenograft, and its biological effects on cellular metabolism, DNA methylation, and transcriptomic using multi-omics approaches. The metabolomics was quantified by liquid-chromatography-mass spectrometry (LC-MS) while epigenomic CpG methylation in parallel with transcriptomic gene expression was studied by next-generation sequencing technologies. UA administration attenuated the growth of transplanted human VCaP-Luc cells in immunodeficient mice. UA regulated several cellular metabolites and metabolism-related signaling pathways including S-adenosylmethionine (SAM), methionine, glucose 6-phosphate, CDP-choline, phosphatidylcholine biosynthesis, glycolysis, and nucleotide sugars metabolism. RNA-seq analyses revealed UA regulated several signaling pathways, including CXCR4 signaling, cancer metastasis signaling, and NRF2-mediated oxidative stress response. Epigenetic reprogramming study with DNA Methyl-seq uncovered a list of differentially methylated regions (DMRs) associated with UA treatment. Transcriptome-DNA methylome correlative analysis uncovered a list of genes, of which changes in gene expression correlated with the promoter CpG methylation status. Altogether, our results suggest that UA regulates metabolic rewiring of metabolism including SAM potentially driving epigenetic CpG methylation reprogramming, and transcriptomic signaling resulting in the overall anticancer chemopreventive effect.


Subject(s)
DNA Methylation/drug effects , Metabolic Networks and Pathways/drug effects , Prostatic Neoplasms/drug therapy , Triterpenes/administration & dosage , Animals , Cell Line, Tumor , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Oxidative Stress/drug effects , Promoter Regions, Genetic/drug effects , Prostatic Neoplasms/genetics , Sequence Analysis, RNA , Triterpenes/pharmacology , Xenograft Model Antitumor Assays , Ursolic Acid
9.
Mol Carcinog ; 60(6): 391-402, 2021 06.
Article in English | MEDLINE | ID: mdl-33848375

ABSTRACT

Epigenetics/epigenomics has been shown to be involved in carcinogenesis. However, how the epigenome would be altered in the transgenic adenocarcinoma of the mouse prostate (TRAMP) cancer model and the effect of cancer chemopreventive phytochemical phenethyl isothiocyanate (PEITC) on the epigenome in TRAMP mice are not known. PEITC has been reported to reduce the risk of many cancers including prostate cancer (PCa). In this study, male TRAMP mice were fed a control diet or diet containing 0.05% PEITC from 8 weeks to 16 weeks. The tumor incidence was reduced in the PEITC diet (0/6) as compared with the control diet (6/7). RNA-sequencing (RNA-seq) analyses on nontumor and tumor prostatic tissues revealed several pathways like cell cycle/Cdc42 signaling, inflammation, and cancer-related signaling, were activated in prostate tissues of TRAMP mice but were reversed or attenuated in TRAMP mice fed with PEITC diet. DNA CpG methyl-seq analyses showed that global methylation patterns of prostate samples from TRAMP mice were hugely different from those of wild-type mice. Dietary PEITC partially reversed the global methylation changes during prostatic carcinogenesis. Integration of RNA-seq and DNA methyl-seq analyses identified a list of genes, including Adgrb1 and Ebf4, with an inverse regulatory relationship between their RNA expression and CpG methylation. In summary, our current study demonstrates that alteration of the global epigenome in TRAMP prostate tumor and PEITC administration suppresses PCa carcinogenesis, impacts global CpG epigenome and transcriptome, and attenuates carcinogenic pathways like cell cycle arrest and inflammation. These results may provide insights and epigenetic markers/targets for PCa prevention and treatment in human PCa patients.


Subject(s)
Anticarcinogenic Agents/pharmacology , DNA Methylation/drug effects , Isothiocyanates/pharmacology , Prostatic Neoplasms/prevention & control , Animals , Epigenome/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Male , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Experimental/genetics , Prostatic Neoplasms/genetics
10.
Cancer Prev Res (Phila) ; 14(2): 151-164, 2021 02.
Article in English | MEDLINE | ID: mdl-33055265

ABSTRACT

Cancer is a complex disease and cancer development takes 10-50 years involving epigenetics. Evidence suggests that approximately 80% of human cancers are linked to environmental factors impinging upon genetics/epigenetics. Because advanced metastasized cancers are resistant to radiotherapy/chemotherapeutic drugs, cancer prevention by relatively nontoxic chemopreventive "epigenetic modifiers" involving epigenetics/epigenomics is logical. Isothiocyanates are relatively nontoxic at low nutritional and even higher pharmacologic doses, with good oral bioavailability, potent antioxidative stress/antiinflammatory activities, possess epigenetic-modifying properties, great anticancer efficacy in many in vitro cell culture and in vivo animal models. This review summarizes the latest advances on the role of epigenetics/epigenomics by isothiocyanates in prevention of skin, colon, lung, breast, and prostate cancers. The exact molecular mechanism how isothiocyanates modify the epigenetic/epigenomic machinery is unclear. We postulate "redox" processes would play important roles. In addition, isothiocyanates sulforaphane and phenethyl isothiocyanate, possess multifaceted molecular mechanisms would be considered as "general" cancer preventive agents not unlike chemotherapeutic agents like platinum-based or taxane-based drugs. Analogous to chemotherapeutic agents, the isothiocyanates would need to be used in combination with other nontoxic chemopreventive phytochemicals or drugs such as NSAIDs, 5-α-reductase/aromatase inhibitors targeting different signaling pathways would be logical for the prevention of progression of tumors to late advanced metastatic states.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Isothiocyanates/therapeutic use , Neoplasms/prevention & control , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Aromatase Inhibitors/pharmacology , Aromatase Inhibitors/therapeutic use , Biological Availability , Disease Models, Animal , Humans , Isothiocyanates/pharmacology , Neoplasms/genetics , Oxidation-Reduction/drug effects
11.
Curr Pharmacol Rep ; 6(3): 56-70, 2020.
Article in English | MEDLINE | ID: mdl-32395418

ABSTRACT

The current pandemic of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has presented unprecedented challenges to the healthcare systems in almost every country around the world. Currently, there are no proven effective vaccines or therapeutic agents against the virus. Current clinical management includes infection prevention and control measures and supportive care including supplemental oxygen and mechanical ventilatory support. Evolving research and clinical data regarding the virologic SARS-CoV-2 suggest a potential list of repurposed drugs with appropriate pharmacological effects and therapeutic efficacies in treating COVID-19 patients. In this review, we will update and summarize the most common and plausible drugs for the treatment of COVID-19 patients. These drugs and therapeutic agents include antiviral agents (remdesivir, hydroxychloroquine, chloroquine, lopinavir, umifenovir, favipiravir, and oseltamivir), and supporting agents (Ascorbic acid, Azithromycin, Corticosteroids, Nitric oxide, IL-6 antagonists), among others. We hope that this review will provide useful and most updated therapeutic drugs to prevent, control, and treat COVID-19 patients until the approval of vaccines and specific drugs targeting SARS-CoV-2.

12.
Cancer Prev Res (Phila) ; 13(8): 673-686, 2020 08.
Article in English | MEDLINE | ID: mdl-32467291

ABSTRACT

Ductal carcinoma in situ (DCIS), which accounts for one out of every five new breast cancer diagnoses, will progress to potentially lethal invasive ductal carcinoma (IDC) in about 50% of cases. Vitamin D compounds have been shown to inhibit progression to IDC in the MCF10DCIS model. This inhibition appears to involve a reduction in the cancer stem cell-like population in MCF10DCIS tumors. To identify genes that are involved in the vitamin D effects, a global transcriptomic analysis was undertaken of MCF10DCIS cells grown in mammosphere cultures, in which cancer stem-like cells grow preferentially and produce colonies by self-renewal and maturation, in the presence and absence of 1α25(OH)2D3 and a vitamin D analog, BXL0124. Using next-generation RNA-sequencing, we found that vitamin D compounds downregulated genes involved in maintenance of breast cancer stem-like cells (e.g., GDF15), epithelial-mesenchymal transition, invasion, and metastasis (e.g., LCN2 and S100A4), and chemoresistance (e.g., NGFR, PPP1R1B, and AGR2), while upregulating genes associated with a basal-like phenotype (e.g., KRT6A and KRT5) and negative regulators of breast tumorigenesis (e.g., EMP1). Gene methylation status was analyzed to determine whether the changes in expression induced by vitamin D compounds occurred via this mechanism. Ingenuity pathway analysis was performed to identify upstream regulators and downstream signaling pathway genes differentially regulated by vitamin D, including TP63 and vitamin D receptor -mediated canonical pathways in particular. This study provides a global profiling of changes in the gene signature of DCIS regulated by vitamin D compounds and possible targets for chemoprevention of DCIS progression to IDC in patients.


Subject(s)
Breast Neoplasms/drug therapy , Carcinoma, Ductal, Breast/prevention & control , Carcinoma, Intraductal, Noninfiltrating/drug therapy , Neoplastic Stem Cells/drug effects , Vitamin D/administration & dosage , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Methylation/drug effects , Datasets as Topic , Disease Progression , Down-Regulation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Neoplastic Stem Cells/pathology , RNA-Seq , Signal Transduction/drug effects , Signal Transduction/genetics , Up-Regulation/drug effects , Vitamin D/analogs & derivatives
13.
Biochem Pharmacol ; 175: 113890, 2020 05.
Article in English | MEDLINE | ID: mdl-32119837

ABSTRACT

Triterpenoids are a powerful group of phytochemicals derived from plant foods and herbs. Many reports have shown that they possess chemopreventive and chemotherapeutic effects not only in cell lines and animal models but also in clinical trials. Because epigenetic changes could potentially occur in the early stages of carcinogenesis preceding genetic mutations, epigenetics are considered promising targets in early interventions against cancer using epigenetic bioactive substances. The biological properties of triterpenoids in cancer prevention and in health have multiple mechanisms, including antioxidant and anti-inflammatory activities, cell cycle regulation, as well as epigenetic/epigenomic regulation. In this review, we will discuss and summarize the latest advances in the study of the pharmacological effects of triterpenoids in cancer chemoprevention and in health, including the epigenetic machinery.


Subject(s)
Anticarcinogenic Agents/pharmacology , Epigenesis, Genetic/drug effects , Neoplasms/genetics , Neoplasms/prevention & control , Phytochemicals/pharmacology , Triterpenes/pharmacology , Anticarcinogenic Agents/chemistry , Cell Line, Tumor , Humans , Phytochemicals/chemistry , Triterpenes/chemistry
14.
Cancer Prev Res (Phila) ; 13(6): 551-562, 2020 06.
Article in English | MEDLINE | ID: mdl-32161072

ABSTRACT

Sulforaphane (SFN), a potent antioxidant and antiinflammatory agent, has been shown to protect against cancers especially at early stages. However, how SFN affects UVB-mediated epigenome/DNA methylome and transcriptome changes in skin photodamage has not been fully assessed. Herein, we investigated the transcriptomic and DNA methylomic changes during tumor initiation, promotion, and progression and its impact and reversal by SFN using next-generation sequencing (NGS) technology. The results show that SFN reduced tumor incidence and tumor number. SFN's protective effects were more dramatic in the early stages than with later stages. Bioinformatic analysis of RNA sequencing (RNA-seq) data shows differential expressed genes and identifies the top canonical pathways related to SFN treatment of UVB-induced different stages of epidermal carcinogenesis. These pathways include p53 signaling, cell cycle: G2-M DNA damage checkpoint regulation, Th1, and Th2 activation pathway, and PTEN signaling pathways. The top upstream regulators related to UVB and SFN treatment as time progressed include dextran sulfate, TP53, NFE2L2 (Nrf2), IFNB1, and IL10RA. Bioinformatic analysis of Methyl-seq data shows several differential methylation regions induced by UVB were attenuated by SFN. These include Notch1, Smad6, Gnai3, and Apc2 Integrative analysis of RNA-seq and DNA-seq/CpG methylome yields a subgroup of genes associated with ultraviolet B (UVB) and SFN treatment. The changes in gene expression were inversely correlated with promoter CpG methylation status. These genes include Pik3cd, Matk, and Adm2 In conclusion, our study provides novel insights on the impact of SFN on the transcriptomic and DNA methylomic of UVB-induced different stages of skin cancer in mice.


Subject(s)
Anticarcinogenic Agents/therapeutic use , DNA Methylation/drug effects , Epigenome/drug effects , Isothiocyanates/therapeutic use , Neoplasms, Radiation-Induced/prevention & control , Skin Neoplasms/prevention & control , Sulfoxides/therapeutic use , Transcriptome/drug effects , Ultraviolet Rays/adverse effects , Acetone/toxicity , Animals , CpG Islands/drug effects , DNA, Neoplasm/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic/genetics , Mice , Mice, Hairless , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/genetics , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , RNA-Seq , Radiation-Sensitizing Agents/toxicity , Random Allocation , Skin Neoplasms/etiology , Skin Neoplasms/genetics
15.
J Pharmacokinet Pharmacodyn ; 47(2): 131-144, 2020 04.
Article in English | MEDLINE | ID: mdl-32020381

ABSTRACT

Curcumin (CUR) is a major component of turmeric Curcuma longa, which is often used in food or as a dietary supplement. The purpose of this preclinical study is to investigate the acute pharmacokinetic and pharmacodynamic (PK/PD) profiles of two commercially marketed CUR products (GNC and Vitamin Shoppe) and a CUR powder from Sigma in female rats. Plasma samples were collected at specific time points and analyzed for CUR and its metabolite curcumin-O-glucuronide. RNA was extracted from leukocytes and analyzed for the expression of Nrf2-mediated antioxidant genes Nrf2, Ho-1, and Nqo1 by qPCR as selected PD markers. CUR PK was characterized by a 2-compartment model (2CM) after intravenous (IV) or oral administrations. Compared to IV CUR, the absolute bioavailability (F) of CUR for GNC (GC) is 0.9%, Vitamin Shoppe (VC) is 0.6% and Sigma (SC) is 3.1%. Pharmacodynamically, all three formulations showed induction of antioxidant Nrf2, Ho-1 and Nqo1 gene expression in rat leucocytes. PK/PD modeling of CUR's effect on antioxidant gene expression was well captured by an indirect response model. Physiologically based PK modeling and simulation using GastroPlus described the observed PK data reasonably well. In summary, our current study shows that the absolute oral bioavailability of the parent CUR was very low for all three formulations. However, despite the low CUR plasma concentrations, all three oral CUR formulations displayed PD response in the induction of Nrf2-mediated antioxidant genes, suggesting the potential of oral CUR contributing to the overall health beneficial effects of oral CUR.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Curcumin/administration & dosage , Curcumin/pharmacokinetics , Administration, Intravenous , Administration, Oral , Animals , Antioxidants/metabolism , Curcuma , Curcumin/analogs & derivatives , Drug Compounding , Female , Glucuronides , Heme Oxygenase-1/metabolism , Leukocytes/drug effects , Leukocytes/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/drug effects , Plant Extracts , Powders , RNA, Messenger/analysis , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
16.
Mol Carcinog ; 59(2): 227-236, 2020 02.
Article in English | MEDLINE | ID: mdl-31820492

ABSTRACT

Colorectal cancer (CRC) is associated with significant morbidity and mortality in the US and worldwide. CRC is the second most common cancer-related death in both men and women globally. Chronic inflammation has been identified as one of the major risk factors of CRC. It may drive genetic and epigenetic/epigenomic alterations, such as DNA methylation, histone modification, and non-coding RNA regulation. Current prevention modalities for CRC are limited and some treatment regimens such as use the nonsteroidal anti-inflammatory drug aspirin may have severe side effects, namely gastrointestinal ulceration and bleeding. Therefore, there is an urgent need of developing alternative strategies. Recently, increasing evidence suggests that several dietary cancer chemopreventive phytochemicals possess anti-inflammation and antioxidative stress activities, and may prevent cancers including CRC. Curcumin (CUR) is the yellow pigment that is found in the rhizomes of turmeric (Curcuma longa). Many studies have demonstrated that CUR exhibit strong anticancer, antioxidative stress, and anti-inflammatory activities by regulating signaling pathways, such as nuclear factor erythroid-2-related factor 2, nuclear factor-κB, and epigenetics/epigenomics pathways of histones modifications, and DNA methylation. In this review, we will discuss the latest evidence in epigenetics/epigenomics alterations by CUR in CRC and their potential contribution in the prevention of CRC.


Subject(s)
Colonic Neoplasms/prevention & control , Curcumin/pharmacology , Epigenesis, Genetic/drug effects , Epigenomics , Inflammation/prevention & control , Antineoplastic Agents/pharmacology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Curcuma/chemistry , Humans , Inflammation/genetics , Inflammation/pathology , Neoplasm Staging , Phytotherapy/methods
17.
AAPS J ; 22(1): 8, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31807911

ABSTRACT

Moringa isothiocyanate (MIC-1) is a bioactive constituent found abundantly in Moringa oleifera which possesses antioxidant and anti-inflammation properties. However, epigenome and transcriptome effects of MIC-1 in kidney mesangial cells challenged with high glucose (HG), a pre-condition for diabetic nephropathy (DN) remain unknown. Herein, we examined the transcriptome gene expression and epigenome DNA methylation in mouse kidney mesangial cells (MES13) using next-generation sequencing (NGS) technology. After HG treatment, epigenome and transcriptome were significantly altered. More importantly, MIC-1 exposure reversed some of the changes caused by HG. Integrative analysis of RNA-Seq data identified 20 canonical pathways showing inverse correlations between HG and MIC-1. These pathways included GNRH signaling, P2Y purigenic receptor signaling pathway, calcium signaling, LPS/IL-1-mediated inhibition of RXR function, and oxidative ethanol degradation III. In terms of alteration of DNA methylation patterns, 173 differentially methylation regions (DMRs) between the HG group and low glucose (LG) group and 149 DMRs between the MIC-1 group and the HG group were found. Several HG related DMRs could be reversed by MIC-1 treatment. Integrative analysis of RNA-Seq and Methyl-Seq data yielded a subset of genes associated with HG and MIC-1, and the gene expression changes may be driven by promoter CpG status. These genes include Col4a2, Tceal3, Ret, and Agt. In summary, our study provides novel insights related to transcriptomic and epigenomic/CpG methylomic alterations in MES13 upon challenged by HG but importantly, MIC-1 treatment reverses some of the transcriptome and epigenome/CpG methylome. These results may provide potential molecular targets and therapeutic strategies for DN.


Subject(s)
Diabetic Nephropathies/drug therapy , Epigenome/drug effects , Isothiocyanates/therapeutic use , Mesangial Cells/drug effects , Rhamnose/analogs & derivatives , Transcriptome/drug effects , Animals , Cell Line , Drug Evaluation, Preclinical , Glucose , Isothiocyanates/pharmacology , Mesangial Cells/metabolism , Mice , Moringa oleifera , Phytotherapy , Reactive Oxygen Species/metabolism , Rhamnose/pharmacology , Rhamnose/therapeutic use , Signal Transduction/drug effects
18.
Chem Res Toxicol ; 32(10): 1977-1988, 2019 10 21.
Article in English | MEDLINE | ID: mdl-31525975

ABSTRACT

Diabetic nephropathy (DN) is a diabetes complication that comes from overactivation of Renin-Angiotensin System, excessive pro-inflammatory factors, reactive oxygen species (ROS) overproduction, and potential epigenetic changes. Tanshinone IIA (TIIA), a diterpene quinone phytochemical, has been shown to possess powerful antioxidant, anti-inflammatory, epigenetics, and protective effects against different diseases including DN by inhibiting ROS induced by high glucose (HG). However, epigenomic and transcriptomic study of DN and the protective effect of TIIA are lacking. In this study, next-generation sequencing of RNA and DNA methylation profiles on the potential underlying mechanisms of a DN model in mouse kidney mesangial mes13 cells challenged with HG and treatment with TIIA were conducted. Bioinformatic analysis coupled with Ingenuity Pathway analysis of RNA-seq was performed, and 1780 genes from HG/LG and 1416 genes from TIIA/HG were significantly altered. Several pro-inflammatory pathways like leukotriene biosynthesis and eicosanoid signaling pathways were activated by HG stimulation, while TIIA treatment would enhance glutathione-mediated detoxification pathway to overcome the excess oxidative stress and inflammation triggered by HG. Combination analysis of RNA-seq and Methyl-seq data sets, DNA methylation, and RNA expression of a list of DN associated genes, Nmu, Fgl2, Glo, and Kcnip2, were found to be altered in HG-induced mes13 DN model, and TIIA treatment would effectively restore the alterations. Taken together, these findings provide novel insights into the understanding of how epigenetic/epigenomic modifications could affect the progression of DN and the potential preventive effect of TIIA in DN.


Subject(s)
Abietanes/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , DNA Methylation/drug effects , Diabetic Nephropathies/drug therapy , Glucose/pharmacology , Transcriptome/drug effects , Abietanes/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cells, Cultured , DNA Methylation/genetics , Diabetic Nephropathies/chemically induced , Disease Models, Animal , Glucose/administration & dosage , Inflammation/drug therapy , Mice , Molecular Structure , Oxidative Stress/drug effects
19.
Mol Carcinog ; 58(10): 1738-1753, 2019 10.
Article in English | MEDLINE | ID: mdl-31237383

ABSTRACT

Nonmelanoma skin cancers (NMSCs) are the most common type of skin cancers. Major risk factors for NMSCs include exposure to ultraviolet (UV) irradiation. Ursolic acid (UA) is a natural triterpenoid enriched in blueberries and herbal medicinal products, and possess anticancer activities. This study focuses on the impact of UA on epigenomic, genomic mechanisms and prevention of UVB-mediated NMSC. CpG methylome and RNA transcriptome alterations of early, promotion and late stages of UA treated on UVB-induced NMSC in SKH-1 hairless mice were conducted using CpG methyl-seq and RNA-seq. Samples were collected at weeks 2, 15, and 25, and integrated bioinformatic analyses were performed to identify key pathways and genes modified by UA against UVB-induced NMSC. Morphologically, UA significantly reduced NMSC tumor volume and tumor number. DNA methylome showed inflammatory pathways IL-8, NF-κB, and Nrf2 pathways were highly involved. Antioxidative stress master regulator Nrf2, cyclin D1, DNA damage, and anti-inflammatory pathways were induced by UA. Nrf2, cyclin D1, TNFrsf1b, and Mybl1 at early (2 weeks) and late (25 weeks) stages were identified and validated by quantitative polymerase chain reaction. In summary, integration of CpG methylome and RNA transcriptome studies show UA alters antioxidative, anti-inflammatory, and anticancer pathways in UVB-induced NMSC carcinogenesis. Particularly, UA appears to drive Nrf2 and its upstream/downstream genes, anti-inflammatory (at early stages) and cell cycle regulatory (both early and late stages) genes, of which might contribute to the overall chemopreventive effects of UVB-induced MNSC. This study may provide potential biomarkers/targets for chemoprevention of early stage of UVB-induced NMSC in human.


Subject(s)
DNA Methylation/genetics , Neoplasms, Radiation-Induced/drug therapy , Skin Neoplasms/drug therapy , Transcriptome/genetics , Animals , Anticarcinogenic Agents/pharmacology , Antioxidants/pharmacology , DNA Damage/drug effects , Disease Models, Animal , Epigenome/drug effects , Humans , Mice , Neoplasm Proteins , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/pathology , Oxidative Stress/drug effects , Signal Transduction/drug effects , Skin/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Transcriptome/radiation effects , Triterpenes/pharmacology , Ultraviolet Rays/adverse effects , Ursolic Acid
20.
Chem Biol Interact ; 309: 108701, 2019 Aug 25.
Article in English | MEDLINE | ID: mdl-31181187

ABSTRACT

Pelargonidin, a well-known natural anthocyanidin found in berries strawberries, blueberries, red radishes and other natural foods, has been found to possess health beneficial effects including anti-cancer effect. Herein, we investigated the effect of pelargonidin on cellular transformation in mouse skin epidermal JB6 (JB6 P+) cells induced by tumor promoter 12-O-tetradecanoylphorbol-13-acetate (TPA). Pelargonidin treatment significantly decreased colony formation and suppressed cell viability of JB6 P+ cells. Pelargonidin also induced the anti-oxidant response element (ARE)-luciferase activation in HepG2-C8 cells overexpressing the ARE-luciferase reporter. Knockdown of nuclear factor E2-related factor 2 (Nrf2) in shNrf2 JB6 P+ cells enhanced TPA-induced colony formation and attenuated pelargonidin's blocking effect. Pelargonidin reduced the protein levels of genes encoding methyltransferases (DNMTs) and histone deacetylases (HDACs). Importantly, pelargonidin decreased the DNA methylation in the Nrf2 promoter region of JB6 P+ cells and increased Nrf2 downstream target genes expression, such as NAD(P)H/quinone oxidoreductase 1 (NQO1) and heme oxygenase-1 (HO-1), involved in cellular protection. In summary, our results showed that pelargonidin blocks TPA-induced cell transformation. The possible molecular mechanisms of its potential anti-cancer effects against neoplastic transformation may be attributed to its activation of Nrf2-ARE signaling pathway and its cytoprotective effect.


Subject(s)
Anthocyanins/pharmacology , DNA Demethylation/drug effects , NF-E2-Related Factor 2/metabolism , Animals , Anthocyanins/chemistry , Antioxidant Response Elements/genetics , Cell Line , Cell Survival/drug effects , DNA-Cytosine Methylases/metabolism , Epidermal Cells/cytology , Epidermal Cells/drug effects , Epidermal Cells/metabolism , Gene Expression/drug effects , Hep G2 Cells , Histone Deacetylases/metabolism , Humans , Mice , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/genetics , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/metabolism , Tetradecanoylphorbol Acetate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...