Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Biomolecules ; 14(1)2024 Jan 14.
Article in English | MEDLINE | ID: mdl-38254705

ABSTRACT

The low bioavailability of most phytochemicals limits their anticancer effects in humans. The present study was designed to test whether combining arctigenin (Arc), a lignan mainly from the seed of Arctium lappa, with green tea (GT) and quercetin (Q) enhances the chemopreventive effect on prostate cancer. We performed in vitro proliferation studies on different cell lines. We observed a strong synergistic anti-proliferative effect of GT+Q+Arc in exposing androgen-sensitive human prostate cancer LNCaP cells. The pre-malignant WPE1-NA22 cell line was more sensitive to this combination. No cytotoxicity was observed in normal prostate epithelial PrEC cells. For an in vivo study, 3-week-old, prostate-specific PTEN (phosphatase and tensin homolog) knockout mice were treated with GT+Q, Arc, GT+Q+Arc, or the control daily until 16 weeks of age. In vivo imaging using prostate-specific membrane antigen (PSMA) probes demonstrated that the prostate tumorigenesis was significantly inhibited by 40% (GT+Q), 60% (Arc at 30 mg/kg bw), and 90% (GT+Q+Arc) compared to the control. A pathological examination showed that all control mice developed invasive prostate adenocarcinoma. In contrast, the primary lesion in the GT+Q and Arc alone groups was high-grade prostatic intraepithelial neoplasia (PIN), with low-grade PIN in the GT+Q+Arc group. The combined effect of GT+Q+Arc was associated with an increased inhibition of the androgen receptor, the PI3K/Akt pathway, Ki67 expression, and angiogenesis. This study demonstrates that combining Arc with GT and Q was highly effective in prostate cancer chemoprevention. These results warrant clinical trials to confirm the efficacy of this combination in humans.


Subject(s)
Furans , Lignans , Prostatic Neoplasms , Animals , Male , Mice , Chemoprevention , Lignans/pharmacology , Lignans/therapeutic use , Mice, Knockout , Phosphatidylinositol 3-Kinases , Prostate/drug effects , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/prevention & control , Quercetin/pharmacology , Quercetin/therapeutic use , Tensins , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Tea
2.
Life Sci ; 312: 121128, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36334805

ABSTRACT

BACKGROUND: The long-term prognosis of breast cancer with metastasis remains extremely poor. Genetic alterations in tumor cells result in cellular heterogeneity, promoting cancer cells invasion and colonization in some organs during the metastatic process. CircRNAs are very promising as critical biological markers and precise diagnoses in identifying disease mechanisms and developing new methods for effective treatment. However, the role of aberrant expression of circRNAs in breast cancer progression remains largely unknown. METHODS: RNase R treatment and quantitative RT-PCR (qRT-PCR) were performed for circRNA detection. Transwell chamber assays were used to examine the chemotactic migration and invasion of breast cancer cells. RESULTS: This study identified and characterized the circRAD54L2 originating from exon 1, 2, 3, and 4 of the RAD54L2 gene. Importantly, we found that circRAD54L2, rather than RAD54L2 linear mRNA, was significantly upregulated in breast cancer cell lines. Furthermore, we found that inhibiting circRAD54L2 expression markedly reduced the invasion, metastasis, and proliferation of breast cancer cells via sponging of the miR-888 family, which downregulated the expression of pyruvate dehydrogenase kinase 1 (PDK1). CONCLUSION: Our results showed that circRAD54L2 could regulate PDK1 expression by sponging the miR-888 family competing for the ceRNA mechanism, indicating that circRAD54L2 may act as an essential upstream regulator and providing further mechanistic evidence to support the notion that circRAD54L2/miR-888s/PDK1 is a promising therapeutic target in the treatment of breast cancer.


Subject(s)
MicroRNAs , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , RNA, Circular , Triple Negative Breast Neoplasms , Humans , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics
3.
Biomolecules ; 12(9)2022 09 16.
Article in English | MEDLINE | ID: mdl-36139145

ABSTRACT

Prostate cancer is one of the leading causes of death for men worldwide. The development of resistance, toxicity, and side effects of conventional therapies have made prostate cancer treatment become more intensive and aggressive. Many phytochemicals isolated from plants have shown to be tumor cytotoxic. In vitro laboratory studies have revealed that natural compounds can affect cancer cell proliferation by modulating many crucial cellular signaling pathways frequently dysregulated in prostate cancer. A multitude of natural compounds have been found to induce cell cycle arrest, promote apoptosis, inhibit cancer cell growth, and suppress angiogenesis. In addition, combinatorial use of natural compounds with hormone and/or chemotherapeutic drugs seems to be a promising strategy to enhance the therapeutic effect in a less toxic manner, as suggested by pre-clinical studies. In this context, we systematically reviewed the currently available literature of naturally occurring compounds isolated from vegetables, fruits, teas, and herbs, with their relevant mechanisms of action in prostate cancer. As there is increasing data on how phytochemicals interfere with diverse molecular pathways in prostate cancer, this review discusses and emphasizes the implicated molecular pathways of cell proliferation, cell cycle control, apoptosis, and autophagy as important processes that control tumor angiogenesis, invasion, and metastasis. In conclusion, the elucidation of the natural compounds' chemical structure-based anti-cancer mechanisms will facilitate drug development and the optimization of drug combinations. Phytochemicals, as anti-cancer agents in the treatment of prostate cancer, can have significant health benefits for humans.


Subject(s)
Antineoplastic Agents , Prostatic Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Hormones , Humans , Male , Neovascularization, Pathologic/drug therapy , Phytochemicals/pharmacology , Phytochemicals/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology
4.
Front Immunol ; 13: 891175, 2022.
Article in English | MEDLINE | ID: mdl-35990668

ABSTRACT

Background: The clinical outcomes of breast cancer (BC) are unpredictable due to the high level of heterogeneity and complex immune status of the tumor microenvironment (TME). When set up, multiple long non-coding RNA (lncRNA) signatures tended to be employed to appraise the prognosis of BC. Nevertheless, predicting immunotherapy responses in BC is still essential. LncRNAs play pivotal roles in cancer development through diverse oncogenic signal pathways. Hence, we attempted to construct an oncogenic signal pathway-based lncRNA signature for forecasting prognosis and immunotherapy response by providing reliable signatures. Methods: We preliminarily retrieved RNA sequencing (RNA-seq) data from The Cancer Genome Atlas (TCGA) database and extracted lncRNA profiles by matching them with GENCODE. Following this, Gene Set Variation Analysis (GSVA) was used to identify the lncRNAs closely associated with 10 oncogenic signaling pathways from the TCGA-BRCA (breast-invasive carcinoma) cohort and was further screened by the least absolute shrinkage and selection operator Cox regression model. Next, an lncRNA signature (OncoSig) was established through the expression level of the final 29 selected lncRNAs. To examine survival differences in the stratification described by the OncoSig, the Kaplan-Meier (KM) survival curve with the log-rank test was operated on four independent cohorts (n = 936). Subsequently, multiple Cox regression was used to investigate the independence of the OncoSig as a prognostic factor. With the concordance index (C-index), the time-dependent receiver operating characteristic was employed to assess the performance of the OncoSig compared to other publicly available lncRNA signatures for BC. In addition, biological differences between the high- and low-risk groups, as portrayed by the OncoSig, were analyzed on the basis of statistical tests. Immune cell infiltration was investigated using gene set enrichment analysis (GSEA) and deconvolution tools (including CIBERSORT and ESTIMATE). The combined effect of the Oncosig and immune checkpoint genes on prognosis and immunotherapy was elucidated through the KM survival curve. Ultimately, a pan-cancer analysis was conducted to attest to the prevalence of the OncoSig. Results: The OncoSig score stratified BC patients into high- and low-risk groups, where the latter manifested a significantly higher survival rate and immune cell infiltration when compared to the former. A multivariate analysis suggested that OncoSig is an independent prognosis predictor for BC patients. In addition, compared to the other four publicly available lncRNA signatures, OncoSig exhibited superior predictive performance (AUC = 0.787, mean C-index = 0.714). The analyses of the OncoSig and immune checkpoint genes clarified that a lower OncoSig score meant significantly longer survival and improved response to immunotherapy. In addition to BC, a high OncoSig score in several other cancers was negatively correlated with survival and immune cell infiltration. Conclusions: Our study established a trustworthy and discriminable prognostic signature for BC patients with similar clinical profiles, thus providing a new perspective in the evaluation of immunotherapy responses. More importantly, this finding can be generalized to be applicable to the vast majority of human cancers.


Subject(s)
Breast Neoplasms , RNA, Long Noncoding , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Carcinogenesis/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Immunotherapy , Prognosis , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Signal Transduction/genetics , Tumor Microenvironment/genetics
5.
Front Oncol ; 12: 916246, 2022.
Article in English | MEDLINE | ID: mdl-35847962

ABSTRACT

Background: The long-term prognosis of HCC (hepatocellular carcinoma) with metastasis remains extremely poor. CircRNAs are promising as critical biological markers in identifying disease mechanisms and developing new effective treatments. However, the role of the aberrant expression of circRNAs in HCC progression remains largely unknown. Methods: CircKIF5B location was investigated by RNA fluorescence in situ hybridization (RNA-FISH). For circRNA determination, RNase R treatment and Real-Time Quantitative RT-PCR (qRT-PCR) were performed. Transwell chamber assays examined the chemotactic migration and invasion of liver cancer cells. Results: This study identified the circRNA circKIF5B originating from exons 1, 2, and 3 of the KIF5B gene. Importantly, we found that circKIF5B circRNA, rather than KIF5B linear mRNA, was notably upregulated in liver cancer cell lines and tissues. Moreover, we found that silencing circKIF5B markedly reduced the proliferation, invasion, and metastasis of liver cancer cells by sponging the miR-192 family, thus decreasing the expression of X-linked inhibitor of apoptosis (XIAP). Conclusion: Our data demonstrate that circKIF5B can regulate XIAP expression by sponging miR-192 and miR-215 competing for the ceRNA mechanism, indicating that circKIF5B may act as an essential upstream regulator and providing mechanistic evidence to support the view that circKIF5B/miR-192s/XIAP is a promising therapeutic target for treating liver cancer.

7.
Int J Mol Sci ; 23(3)2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35163380

ABSTRACT

Exosomes are a class of small membrane-bound extracellular vesicles released by almost all cell types and present in all body fluids. Based on the studies of exosome content and their interactions with recipient cells, exosomes are now thought to mediate "targeted" information transfer. Tumor-derived exosomes (TEX) carry a cargo of molecules different from that of normal cell-derived exosomes. TEX functions to mediate distinct biological effects such as receptor discharge and intercellular cross-talk. The immune system defenses, which may initially restrict tumor progression, are progressively blunted by the broad array of TEX molecules that activate suppressive pathways in different immune cells. Herein, we provide a review of the latest research progress on TEX in the context of tumor-mediated immune suppression and discuss the potential as well as challenges of TEX as a target of immunotherapy.


Subject(s)
Exosomes/metabolism , Immunosuppression Therapy , Neoplasms/metabolism , Animals , Humans , Ligands , MicroRNAs/genetics , MicroRNAs/metabolism , Models, Biological
8.
Adv Sci (Weinh) ; 9(6): e2102303, 2022 02.
Article in English | MEDLINE | ID: mdl-35023320

ABSTRACT

Diabetes is directly related to the risk of breast cancer (BC) occurrence and worsened BC prognosis. Currently, there are no specific treatments for diabetes-associated BC. This paper aims to understand the fundamental mechanisms of diabetes-induced BC progression and to develop personalized treatments. It reports a metabolic reprogramming strategy (MRS) that pharmaceutical induction of glucose import and glycolysis with metformin and NF-κB inhibitor (NF-κBi) while blocking the export of excessive lactate via inhibiting monocarboxylate transporter 4 (MCT4) leads to a metabolic crisis within the cancer cells. It demonstrates that the MRS shifts the metabolism of BC cells toward higher production of lactate, blocks lactate secretion, prompts intracellular acidification and induces significant cytotoxicity. Moreover, a novel MCT4 inhibitor CB-2 has been identified by structure-based virtual screening. A triple combination of metformin, CB-2, and trabectedin, a drug that impedes NF-κB signaling, strongly inhibits BC cells. Compared to normal glucose condition, MRS elicits more potent cancer cell-killing effects under high glucose condition. Animal model studies show that diabetic conditions promote the proliferation and progression of BC xenografts in nude mice and that MRS treatment significantly inhibits HG-induced BC progression. Therefore, inhibition of MCT4 combined with metformin/NF-κBi is a promising cancer therapy, especially for diabetes-associated BC.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Diabetes Mellitus, Experimental/metabolism , Metformin/therapeutic use , Monocarboxylic Acid Transporters/antagonists & inhibitors , Muscle Proteins/antagonists & inhibitors , Trabectedin/therapeutic use , Animals , Antineoplastic Agents, Alkylating/metabolism , Antineoplastic Agents, Alkylating/therapeutic use , Breast Neoplasms/complications , Diabetes Mellitus, Experimental/complications , Disease Models, Animal , Female , Glucose/metabolism , Glycolysis/drug effects , Humans , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/therapeutic use , Lactic Acid/metabolism , Metformin/metabolism , Mice , Prognosis , Trabectedin/metabolism
9.
J Cancer Sci Clin Ther ; 5(1): 134-153, 2021 Mar 18.
Article in English | MEDLINE | ID: mdl-34950877

ABSTRACT

TP53 gene is often mutated in gastric cancer (GC), nonetheless its relationship with clinicopathological characteristics and prognosis is still unclear. Here, we sought to ascertain the difference in clinical phenotypes between TP53 wild-type and mutant tumors in confirmed gastric cancer patients. To this end, we analyzed TP53 mutation status of 415 TCGA GC patients in relation to their clinical and pathological features as well as prognosis. Longrank Test showed that the survival rate of gastric cancer patients with TP53 WT was significantly lower than that of TP53 mut. Compared with TP53 mut gastric cancer patients with low mRNA expression, TP53 WT patients with low mRNA expression have lower overall survival rate. The death risk of TP53 WT gastric cancer patients is 1.395 times that of TP53 mut gastric cancer patients. The death risk of TP53 mut gastric cancer patients is not related to age, and advanced age is not a risk factor. However, the death risk of TP53 WT patients with gastric cancer increases with age, and the death risk of patients over 70 years old is 1.899 times that of patients under 60 years old. These results suggest that the prognosis of elderly gastric cancer patients with TP53 WT is worse. CONCLUSION: our results indicate that the status of TP53 mutation in GC is significantly correlated with clinical or molecular categories and that the prognosis of GC patients with WT TP53 is worse than that of patients with mutant TP53. Therefore, our data emphasize the importance of distinguishing TP53 WT to predict poor overall survival and relapse-free survival in patients with GC.

10.
Cell Death Dis ; 11(12): 1052, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33311440

ABSTRACT

The abnormal PI3K/AKT/mTOR pathway is one of the most common genomic abnormalities in breast cancers including triple-negative breast cancer (TNBC), and pharmacologic inhibition of these aberrations has shown activity in TNBC patients. Here, we designed and identified a small-molecule Comp34 that suppresses both AKT and mTOR protein expression and exhibits robust cytotoxicity towards TNBC cells but not nontumorigenic normal breast epithelial cells. Mechanically, long noncoding RNA (lncRNA) AL354740.1-204 (also named as NUDT3-AS4) acts as a microRNA sponge to compete with AKT1/mTOR mRNAs for binding to miR-99s, leading to decrease in degradation of AKT1/mTOR mRNAs and subsequent increase in AKT1/mTOR protein expression. Inhibition of lncRNA-NUDT3-AS4 and suppression of the NUDT3-AS4/miR-99s association contribute to Comp34-affected biologic pathways. In addition, Comp34 alone is effective in cells with secondary resistance to rapamycin, the best-known inhibitor of mTOR, and displays a greater in vivo antitumor efficacy and lower toxicity than rapamycin in TNBC xenografted models. In conclusion, NUDT3-AS4 may play a proproliferative role in TNBC and be considered a relevant therapeutic target, and Comp34 presents promising activity as a single agent to inhibit TNBC through regulation of NUDT3-AS4 and miR-99s.


Subject(s)
Antineoplastic Agents/therapeutic use , Curcumin/therapeutic use , RNA, Long Noncoding/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Antineoplastic Agents/pharmacology , Base Sequence , Cell Line, Tumor , Cell Proliferation/drug effects , Curcumin/chemistry , Curcumin/pharmacology , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Long Noncoding/genetics , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Transcription, Genetic/drug effects
11.
Theranostics ; 10(20): 9230-9248, 2020.
Article in English | MEDLINE | ID: mdl-32802189

ABSTRACT

Rationale: Skeletal muscle insulin resistance is detectable before type 2 diabetes is diagnosed. Exposure to di(2-ethylhexyl) phthalate (DEHP), a typical environmental endocrine-disrupting chemical, is a novel risk factor for insulin resistance and type 2 diabetes. This study aimed to explore insulin signaling regulatory pathway in skeletal muscle of the DEHP-induced insulin-resistant mice and to investigate potential therapeutic strategies for treating insulin resistance. Methods: C57BL/6J male mice were exposed to 2 mg/kg/day DEHP for 15 weeks. Whole-body glucose homeostasis, oxidative stress and deregulated miRNA-mediated molecular transduction in skeletal muscle were examined. microRNA (miRNA) interventions based on lentiviruses and adeno-associated viruses 9 (AAV9) were performed. Results: Dnmt3a-dependent promoter methylation and lncRNA Malat1-related sponge functions cooperatively downregulated miR-17 in DEHP-exposed skeletal muscle cells. DEHP suppressed miR-17 to disrupt the Keap1-Nrf2 redox system and to activate oxidative stress-responsive Txnip in skeletal muscle. Oxidative stress upregulated miR-200a, which directly targets the 3'UTR of Insr and Irs1, leading to hindered insulin signaling and impaired insulin-dependent glucose uptake in skeletal muscle, ultimately promoting the development of insulin resistance. AAV9-induced overexpression of miR-17 and lentivirus-mediated silencing of miR-200a in skeletal muscle ameliorated whole-body insulin resistance in DEHP-exposed mice. Conclusions: The miR-17/Keap1-Nrf2/miR-200a axis contributed to DEHP-induced insulin resistance. miR-17 is a positive regulator, whereas miR-200a is a negative regulator of insulin signaling in skeletal muscle, and both miRNAs have the potential to become therapeutic targets for preventing and treating insulin resistance or type 2 diabetes.


Subject(s)
Epigenetic Repression/genetics , Insulin Resistance/genetics , Kelch-Like ECH-Associated Protein 1/genetics , MicroRNAs/genetics , Muscle, Skeletal/metabolism , NF-E2-Related Factor 2/genetics , Animals , Cell Line , Diabetes Mellitus, Type 2/metabolism , Down-Regulation/genetics , Insulin/genetics , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , Oxidative Stress/genetics , Phthalic Acids/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics
12.
Cell Commun Signal ; 18(1): 82, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32471499

ABSTRACT

Chemokines are a family of small cytokines, which guide a variety of immune/inflammatory cells to the site of tumor in tumorigenesis. A dysregulated expression of chemokines is implicated in different types of cancer including prostate cancer. The progression and metastasis of prostate cancer involve a complex network of chemokines that regulate the recruitment and trafficking of immune cells. The chemokine CCL2 and its main receptor CCR2 have been receiving particular interest on their roles in cancer pathogenesis. The up-regulation of CCL2/CCR2 and varied immune conditions in prostate cancer, are associated with cancer advancement, metastasis, and relapse. Here we reviewed recent findings, which link CCL2/CCR2 to the inflammation and cancer pathogenesis, and discussed the therapeutic potential of CCL2/CCR2 axis in cancer treatment based on results from our group and other investigators, with a major focus on prostate cancer. Video Abstract.


Subject(s)
Chemokine CCL2/physiology , Inflammation/metabolism , Prostatic Neoplasms/metabolism , Receptors, CCR2/physiology , Animals , Humans , Male
13.
Biomed Pharmacother ; 125: 109956, 2020 May.
Article in English | MEDLINE | ID: mdl-32006900

ABSTRACT

A greater understanding of factors causing cancer initiation, progression and evolution is of paramount importance. Among them, the serine/threonine phosphatase PPM1D, also referred to as wild-type p53-induced phosphatase 1 (Wip1) or protein phosphatase 2C delta (PP2Cδ), is emerging as an important oncoprotein due to its negative regulation on a number of crucial cancer suppressor pathways. Initially identified as a p53-regulated gene, PPM1D has been afterwards found amplified and more recently mutated in many human cancers such as breast cancer. The latest progress in this field further reveals that selective inhibition of PPM1D to delay tumor onset or reduce tumor burden represents a promising anti-cancer strategy. Here, we review the advances in the studies of the PPM1D activity and its relevance to various cancers, and recent progress in development of PPM1D inhibitors and discuss their potential application in cancer therapy. Consecutive research on PPM1D and its relationship with cancer is essential, as it ultimately contributes to the etiology and treatment of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , Protein Phosphatase 2C/antagonists & inhibitors , Protein Phosphatase 2C/metabolism , Antineoplastic Agents/chemistry , Molecular Structure , Neoplasms/drug therapy , Protein Phosphatase 2C/genetics
14.
Sci Rep ; 10(1): 1403, 2020 Jan 29.
Article in English | MEDLINE | ID: mdl-31996731

ABSTRACT

This study investigated the inhibitory effect of arctigenin, a novel anti-inflammatory lignan, on prostate cancer in obese conditions both in vitro and in vivo. In vitro obese models were established by co-culture of mouse adipocytes 3T3-L1 with androgen-sensitive LNCaP human prostate cancer cells, or by culturing LNCaP cells in adipocytes-conditioned medium. Arctigenin significantly inhibited LNCaP proliferation, along with decreased androgen receptor (AR) and increased Nkx3.1 cellular expression. Male severe combined immunodeficiency mice were subcutaneously implanted with human prostate cancer LAPC-4 xenograft tumors for in vivo study. Mice were fed high-fat (HF) diet and orally given arctigenin at 50 mg/kg body weight daily or vehicle control for 6 weeks. Tumor bearing HF control mice showed a significant increase in serum free fatty acids (FFAs) and decrease in subcutaneous/peritoneal fat depots compared to non-tumor bearing control mice. Arctigenin intervention significantly reduced tumor growth by 45%, associated with decreased circulating FFAs and adipokines/cytokines including IGF-1, VEGF, and MCP-1, along with decreased AR, Ki67, and microvessel density and increased Nkx3.1 expression in tumors. These results indicate the strong ability of arctigenin to co-target obesity and tumor itself in inhibition of prostate tumor growth at a lower concentration compared to most phytochemicals.


Subject(s)
Adipose Tissue/drug effects , Furans/administration & dosage , Lignans/administration & dosage , Obesity/drug therapy , Prostatic Neoplasms/drug therapy , Tumor Burden/drug effects , Adipocytes/drug effects , Adipocytes/metabolism , Adipokines/blood , Adipokines/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Administration, Oral , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cytokines/blood , Cytokines/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Homeodomain Proteins/metabolism , Humans , Male , Mice , Obesity/etiology , Obesity/metabolism , Prostate/pathology , Prostatic Neoplasms/complications , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
15.
Biosci Rep ; 39(12)2019 12 20.
Article in English | MEDLINE | ID: mdl-31710081

ABSTRACT

Mitochondrial reactive oxygen species (ROS) cause Ca2+ release from the endoplasmic reticulum (ER) via ryanodine receptors (RyRs) in pulmonary artery smooth muscle cells (PASMCs), playing an essential role in hypoxic pulmonary vasoconstriction (HPV). Here we tested a novel hypothesis that hypoxia-induced RyR-mediated Ca2+ release may, in turn, promote mitochondrial ROS generation contributing to hypoxic cellular responses in PASMCs. Our data reveal that application of caffeine to elevate intracellular Ca2+ concentration ([Ca2+]i) by activating RyRs results in a significant increase in ROS production in cytosol and mitochondria of PASMCs. Norepinephrine to increase [Ca2+]i due to the opening of inositol 1,4,5-triphosphate receptors (IP3Rs) produces similar effects. Exogenous Ca2+ significantly increases mitochondrial-derived ROS generation as well. Ru360 also inhibits the hypoxic ROS production. The RyR antagonist tetracaine or RyR2 gene knockout (KO) suppresses hypoxia-induced responses as well. Inhibition of mitochondrial Ca2+ uptake with Ru360 eliminates N- and Ca2+-induced responses. RISP KD abolishes the hypoxia-induced ROS production in mitochondria of PASMCs. Rieske iron-sulfur protein (RISP) gene knockdown (KD) blocks caffeine- or NE-induced ROS production. Taken together, these findings have further demonstrated that ER Ca2+ release causes mitochondrial Ca2+ uptake and RISP-mediated ROS production; this novel local ER/mitochondrion communication-elicited, Ca2+-mediated, RISP-dependent ROS production may play a significant role in hypoxic cellular responses in PASMCs.


Subject(s)
Calcium/metabolism , Electron Transport Complex III/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/metabolism , Reactive Oxygen Species/metabolism , Animals , Electron Transport Complex III/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Male , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/genetics , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Pulmonary Artery/pathology
16.
Sci Adv ; 5(10): eaaw8417, 2019 10.
Article in English | MEDLINE | ID: mdl-31663018

ABSTRACT

Although nuclear type 2C protein phosphatase (PP2Cδ) has been demonstrated to be pro-oncogenic with an important role in tumorigenesis, the underlying mechanisms that link aberrant PP2Cδ levels with cancer development remain elusive. Here, we found that aberrant PP2Cδ activity decreases p53 acetylation and its transcriptional activity and suppresses doxorubicin-induced cell apoptosis. Mechanistically, we show that BRCA1 facilitates p300-mediated p53 acetylation by complexing with these two proteins and that S1423/1524 phosphorylation is indispensable for this regulatory process. PP2Cδ, via dephosphorylation of ATM, suppresses DNA damage-induced BRCA1 phosphorylation, leading to inhibition of p300-mediated p53 acetylation. Furthermore, PP2Cδ levels correlate with histological grade and are inversely associated with BRCA1 phosphorylation and p53 acetylation in breast cancer specimens. C23, our newly developed PP2Cδ inhibitor, promotes the anticancer effect of doxorubicin in MCF-7 xenograft-bearing nude mice. Together, our data indicate that PP2Cδ impairs p53 acetylation and DNA damage response by compromising BRCA1 function.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , BRCA1 Protein/genetics , Breast Neoplasms/genetics , DNA Damage/genetics , E1A-Associated p300 Protein/genetics , Protein Phosphatase 2C/genetics , Signal Transduction/genetics , Tumor Suppressor Protein p53/genetics , Acetylation , Animals , Apoptosis/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Female , Humans , MCF-7 Cells , Mice , Mice, Nude , Phosphorylation/genetics
17.
Breast Cancer Res ; 21(1): 29, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30791936

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive subset of breast carcinomas that lack expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER2). Unlike other breast cancer subtypes, targeted therapy is presently unavailable for patients with TNBC. In spite of initial responses to chemotherapy, drug resistance tends to develop rapidly and the prognosis of metastatic TNBC is poor. Hence, there is an urgent need for novel-targeted treatment methods or development of safe and effective alternatives with recognized mechanism(s) of action. AMP-activated protein kinase (AMPK), an energy sensor, can regulate protein and lipid metabolism responding to alterations in energy supply. In the past 10 years, interest in AMPK has increased widely since it appeared as an attractive targeting molecule for cancer therapy. There has been a deep understanding of the possible role of abnormal AMPK signaling pathways in the regulation of growth and survival and the development of drug resistance in TNBC. The increasing popularity of using AMPK regulators for TNBC-targeted therapy is supported by a considerable development in ascertaining the molecular pathways implicated. This review highlights the available evidence for AMPK-targeted anti-TNBC activity of various agents or treatment strategies, with special attention placed on recent preclinical and clinical advances in the manipulation of AMPK in TNBC. The elaborative analysis of these AMPK-related signaling pathways will have a noteworthy impact on the development of AMPK regulators, resulting in efficacious treatments for this lethal disease.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents/therapeutic use , Enzyme Activators/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Triple Negative Breast Neoplasms/therapy , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/genetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Enzyme Activators/pharmacology , Female , Genetic Therapy/methods , Humans , Metabolic Networks and Pathways/drug effects , MicroRNAs/administration & dosage , MicroRNAs/genetics , Molecular Targeted Therapy/methods , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/pathology
18.
Medicine (Baltimore) ; 98(3): e13959, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30653100

ABSTRACT

BACKGROUND: The value of layer-specific two-dimensional speckle tracking echocardiography (LS2D-STE) for evaluating viable myocardium (VM) in patients with acute myocardial infarction (AMI) was unclear, this study provides new insights into it and to make a comparison with dualisotope simultaneous acquisition single photon emission computed tomography ( DISA-SPECT). METHODS: Forty hospitalized patients with AMI and left ventricular systolic dysfunction (left ventricular ejection fraction <50%) underwent LS2D-STE and DISA-SPECT before percutaneous coronary intervention (PCI). The longitudinal, circumferential, and radial peak systolic strains and the peak systolic strain rates of 3 myocardiallayers (endocardium, mid-myocardium, and epicardium), as well as the total wall thickness, were determined by LS2D-STE. Routine echocardiography was followedup at 1, 3, 6 months after PCI, with the improvement of the wall motion as the goldenstandard for evaluating VM. RESULTS: The sensitivity, specificity and accuracy of DISA-SPECT for evaluating VM were 82.1%, 74.3%, and 79.3%, respectively. Among the layer-specific parameters, only endocardial (endo-) longitudinal strain (LS) and endo- longitudinal strain rate (LSr) were used as independent parameters for evaluating VM (P < .05), and the sensitivity, specificity and accuracy of endo-LS and endo-LSr in evaluation of VM were 77.1%, 65.4%, and 72.9% vs 72.9%, 65.4%, and 69.7%. Endo-LS and endo-LSr were superior to total wall thickness LS and LSr (AUC endo-LS 0.767 vs total-LS 0.669; endo-LSr 0.743 vs total-LSr 0.682). The parallel test and the serial test of combination of endo-LS and endo-LSr showed similar sensitivity, specificity and accuracy to DISA-SPECT (P > .05). CONCLUSION: The endo-LS and endo-LSr analysis of LS2D-STE can evaluate the VM well, and its sensitivity, specificity and accuracy in detection of VM are similar to those of DISA-SPECT, resulting in LS2D-STE being a good option for the assessment of VM.


Subject(s)
Echocardiography/instrumentation , Myocardial Infarction/diagnostic imaging , Myocardium/cytology , Percutaneous Coronary Intervention/methods , Tissue Survival/physiology , Acute Disease , Aged , Endocardium/diagnostic imaging , Endocardium/pathology , Female , Hospitalization/statistics & numerical data , Humans , Male , Middle Aged , Myocardial Infarction/pathology , Myocardial Infarction/surgery , Myocardium/pathology , Tomography, Emission-Computed, Single-Photon/methods , Ventricular Dysfunction, Left/physiopathology
19.
Antioxid Redox Signal ; 30(17): 1983-1998, 2019 06 10.
Article in English | MEDLINE | ID: mdl-29808718

ABSTRACT

Aims: Epidemiologic evidence indicates that diabetes may increase risk of breast cancer (BC) and mortality in patients with cancer. The pathophysiological relationships between diabetes and cancer are not fully understood, and personalized treatments for diabetes-associated BC are urgently needed. Results: We observed that high glucose (HG), via activation of nuclear phosphatase PP2Cδ, suppresses p53 function, and consequently promotes BC cell proliferation, migration, and invasion. PP2Cδ expression is higher in tumor tissues from BC patients with hyperglycemia than those with normoglycemia. The mechanisms underlying HG stimulation of PP2Cδ involve classical/novel protein kinase-C (PKC) activation and GSK3ß phosphorylation. Reactive oxygen species (ROS)/NF-κB pathway also mediates HG induction of PP2Cδ. Furthermore, we identified a 1,5-diheteroarylpenta-1,4-dien-3-one (Compound 23, or C23) as a novel potent PP2Cδ inhibitor with a striking cytotoxicity on MCF-7 cells through cell-based screening assay for growth inhibition and activity of a group of curcumin mimics. Beside directly inhibiting PP2Cδ activity, C23 blocks HG induction of PP2Cδ expression via heat shock protein 27 (HSP27) induction and subsequent ablation of ROS/NF-κB activation. C23 can thus significantly block HG-triggered inhibition of p53 activity, leading to the inhibition of cancer cell proliferation, migration, and invasion. In addition, hyperglycemia promotes BC development in diabetic nude mice, and C23 inhibits the xenografted BC tumor growth. Conclusions and Innovation: Our findings elucidate mechanisms that may have contributed to diabetes-associated BC progression, and provide the first evidence to support the possible alternative therapeutic approach to BC patients with diabetes. Antioxid. Redox Signal. 30, 1983-1998.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Curcumin/pharmacology , Enzyme Inhibitors/pharmacology , Glucose/metabolism , Protein Phosphatase 2C/antagonists & inhibitors , Acetylation , Animals , Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Cell Line, Tumor , Curcumin/analogs & derivatives , Curcumin/chemistry , Disease Models, Animal , Disease Progression , Enzyme Inhibitors/chemistry , Female , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Hyperglycemia , Mice , Models, Molecular , NF-kappa B/metabolism , Phosphorylation , Protein Phosphatase 2C/chemistry , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
20.
Cancer Manag Res ; 10: 4653-4667, 2018.
Article in English | MEDLINE | ID: mdl-30464589

ABSTRACT

Breast cancer has a high incidence worldwide. The results of substantial studis reveal that inflammation plays an important role in the initiation, development, and aggressiveness of many malignancies. The use of celecoxib, a novel NSAID, is repetitively associated with the reduced risk of the occurrence and progression of a number of types of cancer, particularly breast cancer. This observation is also substantiated by various meta-analyses. Clinical trials have been implemented on integration treatment of celecoxib and shown encouraging results. Celecoxib could be treated as a potential candidate for antitumor agent. There are, nonetheless, some unaddressed questions concerning the precise mechanism underlying the anticancer effect of celecoxib as well as its activity against different types of cancer. In this review, we discuss different mechanisms of anticancer effect of celecoxib as well as preclinical/clinical results signifying this beneficial effect.

SELECTION OF CITATIONS
SEARCH DETAIL