Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 29(7): 1305-1316, 2023 04 03.
Article in English | MEDLINE | ID: mdl-36648507

ABSTRACT

PURPOSE: Mutations of the isocitrate dehydrogenase (IDH) gene are common genetic mutations in human malignancies. Increasing evidence indicates that IDH mutations play critical roles in malignant transformation and progression. However, the therapeutic options for IDH-mutated cancers remain limited. In this study, the investigation of patient cohorts revealed that the PI3K/protein kinase B (AKT) signaling pathways were enhanced in IDH-mutated cancer cells. EXPERIMENTAL DESIGN: In this study, we investigated the gene expression profile in IDH-mutated cells using RNA sequencing after the depletion of AKT. Gene set enrichment analysis (GSEA) and pathway enrichment analysis were used to discover altered molecular pathways due to AKT depletion. We further investigated the therapeutic effect of the AKT inhibitor, ipatasertib (Ipa), combined with temozolomide (TMZ) in cell lines and preclinical animal models. RESULTS: GSEA and pathway enrichment analysis indicated that the PI3K/AKT pathway significantly correlated with Nrf2-guided gene expression and ferroptosis-related pathways. Mechanistically, AKT suppresses the activity of GSK3ß and stabilizes Nrf2. Moreover, inhibition of AKT activity with Ipa synergizes with the genotoxic agent TMZ, leading to overwhelming ferroptotic cell death in IDH-mutated cancer cells. The preclinical animal model confirmed that combining Ipa and TMZ treatment prolonged survival. CONCLUSIONS: Our findings highlighted AKT/Nrf2 pathways as a potential synthetic lethality target for IDH-mutated cancers.


Subject(s)
Brain Neoplasms , Ferroptosis , Glioma , Animals , Humans , Proto-Oncogene Proteins c-akt/metabolism , Isocitrate Dehydrogenase/genetics , NF-E2-Related Factor 2/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Ferroptosis/genetics , Cell Line, Tumor , Glioma/drug therapy , Glioma/genetics , Glioma/pathology , Temozolomide/pharmacology , Temozolomide/therapeutic use , Mutation , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism
2.
J Exp Clin Cancer Res ; 41(1): 235, 2022 Aug 03.
Article in English | MEDLINE | ID: mdl-35918767

ABSTRACT

BACKGROUND: Radiation therapy (RT) with androgen deprivation therapy (ADT) is an effective therapy to suppress the locally advanced prostate cancer (PCa). However, we unexpectedly found that RT could also induce the androgen receptor splice variant 7 (ARv7) expression to decrease the radiosensitivity. METHODS: The study was designed to target ARv7 expression with Quercetin or ARv7-shRNA that leads to enhancing and increasing the radiation sensitivity to better suppress the PCa that involved the modulation of the circNHS/miR-512-5p/XRCC5 signaling. RESULTS: Mechanism studies revealed that RT-induced ARv7 may function via altering the circNHS/miR-512-5p/XRCC5 signaling to decrease the radiosensitivity. Results from preclinical studies using multiple in vitro cell lines and in vivo mouse models concluded that combining RT with the small molecule of Quercetin to target full-length AR and ARv7 could lead to better efficacy to suppress PCa progression. CONCLUSION: Together, these results suggest that ARv7 may play key roles to alter the PCa radiosensitivity, and targeting this newly identified ARv7 mediated circNHS/miR-512-5p/XRCC5 signaling with Quercetin may help physicians to develop a novel RT to better suppress the progression of PCa.


Subject(s)
MicroRNAs , Prostatic Neoplasms , Androgen Antagonists , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Ku Autoantigen/genetics , Ku Autoantigen/metabolism , Male , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/radiotherapy , Quercetin/pharmacology , Radiation Tolerance , Receptors, Androgen/metabolism
3.
Cells ; 10(9)2021 09 07.
Article in English | MEDLINE | ID: mdl-34571995

ABSTRACT

Isocitrate dehydrogenase (IDH) mutations are common genetic abnormalities in glioma, which result in the accumulation of an "oncometabolite", D-2-hydroxyglutarate (D-2-HG). Abnormally elevated D-2-HG levels result in a distinctive pattern in cancer biology, through competitively inhibiting α-ketoglutarate (α-KG)/Fe(II)-dependent dioxgenases (α-KGDDs). Recent studies have revealed that D-2-HG affects DNA/histone methylation, hypoxia signaling, DNA repair, and redox homeostasis, which impacts the oncogenesis of IDH-mutated cancers. In this review, we will discuss the current understanding of D-2-HG in cancer biology, as well as the emerging opportunities in therapeutics in IDH-mutated glioma.


Subject(s)
Glioma/metabolism , Glutarates/metabolism , Isocitrate Dehydrogenase/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Brain Neoplasms/genetics , Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , DNA Methylation , Glioma/genetics , Glioma/physiopathology , Glutarates/adverse effects , Humans , Hypoxia/metabolism , Isocitrate Dehydrogenase/metabolism , Ketoglutaric Acids/metabolism , Mutation , Signal Transduction/physiology
4.
Pharmacol Ther ; 228: 107922, 2021 12.
Article in English | MEDLINE | ID: mdl-34171339

ABSTRACT

Glioma is one of the most common and lethal brain tumors. Surgical resection followed by radiotherapy plus chemotherapy is the current standard of care for patients with glioma. The existence of resistance to genotoxic therapy, as well as the nature of tumor heterogeneity greatly limits the efficacy of glioma therapy. DNA damage repair pathways play essential roles in many aspects of glioma biology such as cancer progression, therapy resistance, and tumor relapse. O6-methylguanine-DNA methyltransferase (MGMT) repairs the cytotoxic DNA lesion generated by temozolomide (TMZ), considered as the main mechanism of drug resistance. In addition, mismatch repair, base excision repair, and homologous recombination DNA repair also play pivotal roles in treatment resistance as well. Furthermore, cellular mechanisms, such as cancer stem cells, evasion from apoptosis, and metabolic reprogramming, also contribute to TMZ resistance in gliomas. Investigations over the past two decades have revealed comprehensive mechanisms of glioma therapy resistance, which has led to the development of novel therapeutic strategies and targeting molecules.


Subject(s)
Brain Neoplasms , Glioma , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , DNA Damage/drug effects , Glioma/drug therapy , Glioma/genetics , Humans
5.
J Exp Clin Cancer Res ; 40(1): 3, 2021 Jan 04.
Article in English | MEDLINE | ID: mdl-33390173

ABSTRACT

BACKGROUND: Early studies indicated that ASC-J9®, an androgen receptor (AR) degradation enhancer, could suppress the prostate cancer (PCa) progression. Here we found ASC-J9® could also suppress the PCa progression via an AR-independent mechanism, which might involve modulating the tumor suppressor ATF3 expression. METHODS: The lentiviral system was used to modify gene expression in C4-2, CWR22Rv1 and PC-3 cells. Western blot and Immunohistochemistry were used to detect protein expression. MTT and Transwell assays were used to test the proliferation and invasion ability. RESULTS: ASC-J9® can suppress PCa cell proliferation and invasion in both PCa C4-2 and CWR22Rv1 cells via altering the ATF3 expression. Further mechanistic studies reveal that ASC-J9® can increase the ATF3 expression via decreasing Glutamate-cysteine ligase catalytic (GCLC) subunit expression, which can then lead to decrease the PTK2 expression. Human clinical studies further linked the ATF3 expression to the PCa progression. Preclinical studies using in vivo mouse model also proved ASC-J9® could suppress AR-independent PCa cell invasion, which could be reversed after suppressing ATF3. CONCLUSIONS: ASC-J9® can function via altering ATF3/PTK2 signaling to suppress the PCa progression in an AR-independent manner.


Subject(s)
Activating Transcription Factor 3/metabolism , Curcumin/analogs & derivatives , Focal Adhesion Kinase 1/metabolism , Prostatic Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Curcumin/pharmacology , Humans , Male , Mice , Mice, Nude , Neoplasm Invasiveness , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
6.
Cell Death Dis ; 11(11): 942, 2020 11 02.
Article in English | MEDLINE | ID: mdl-33139720

ABSTRACT

The FDA-approved anti-androgen Enzalutamide (Enz) has been used successfully as the last line therapy to extend castration-resistant prostate cancer (CRPC) patients' survival by an extra 4.8 months. However, CRPC patients eventually develop Enz-resistance that may involve the induction of the androgen receptor (AR) splicing variant ARv7. Here we found that Cisplatin (Cis) or Carboplatin, currently used in chemotherapy/radiation therapy to suppress tumor progression, could restore the Enz sensitivity in multiple Enz-resistant (EnzR) CRPC cells via directly degrading/suppressing the ARv7. Combining Cis or Carboplatin with Enz therapy can also delay the development of Enz-resistance in CRPC C4-2 cells. Mechanism dissection found that Cis or Carboplatin might decrease the ARv7 expression via multiple mechanisms including targeting the lncRNA-Malat1/SF2 RNA splicing complex and increasing ARv7 degradation via altering ubiquitination. Preclinical studies using in vivo mouse model with implanted EnzR1-C4-2 cells also demonstrated that Cis plus Enz therapy resulted in better suppression of EnzR CRPC progression than Enz treatment alone. These results not only unveil the previously unrecognized Cis mechanism to degrade ARv7 via targeting the Malat1/SF2 complex and ubiquitination signals, it may also provide a novel and ready therapy to further suppress the EnzR CRPC progression in the near future.


Subject(s)
Alternative Splicing , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzamides/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Nitriles/pharmacology , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/metabolism , Animals , Apoptosis , Carboplatin/administration & dosage , Cell Proliferation , Cisplatin/administration & dosage , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Androgen/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Aging (Albany NY) ; 12(17): 17694-17712, 2020 Sep 10.
Article in English | MEDLINE | ID: mdl-32920545

ABSTRACT

Anti-androgen therapy with Enzalutamide (Enz) has been used as a therapy for castration resistant prostate cancer (CRPC) patients after development of resistance to chemotherapy with Docetaxel (Doc). The potential impacts of Doc-chemotherapy on the subsequent Enz treatment, however, remain unclear. Here we found the overall survival rate of patients that received Enz was significantly less in patients that received prior Doc-chemotherapy than those who had not. In vitro studies from 3 established Doc resistant CRPC (DocRPC) cell lines are consistent with the clinical findings showing DocRPC patients had decreased Enz-sensitivity as well as accelerated development of Enz-resistance via enhanced androgen receptor (AR) splicing variant 7 (ARv7) expression. Mechanism dissection found that Doc treatment might increase the generation of ARv7 via altering the MALAT1-SF2 RNA splicing complex. Preclinical studies using in vivo mouse models and in vitro cell lines proved that targeting the MALAT1/SF2/ARv7 axis with small molecules, including siMALAT1, shSF2, and shARv7 or ARv7 degradation enhancers: Cisplatin or ASC-J9®, can restore/increase the Enz sensitivity to further suppress DocRPC cell growth. Therefore, combined therapy of Doc-chemotherapy with anti-ARv7 therapy, including Cisplatin or ASC-J9®, may be developed to increase the efficacy of Enz to further suppress DocRPC in patients.

8.
Biomedicines ; 8(9)2020 Aug 20.
Article in English | MEDLINE | ID: mdl-32825279

ABSTRACT

Mutations in isocitrate dehydrogenase (IDH) are commonly observed in lower-grade glioma and secondary glioblastomas. IDH mutants confer a neomorphic enzyme activity that converts α-ketoglutarate to an oncometabolite D-2-hydroxyglutarate, which impacts cellular epigenetics and metabolism. IDH mutation establishes distinctive patterns in metabolism, cancer biology, and the therapeutic sensitivity of glioma. Thus, a deeper understanding of the roles of IDH mutations is of great value to improve the therapeutic efficacy of glioma and other malignancies that share similar genetic characteristics. In this review, we focused on the genetics, biochemistry, and clinical impacts of IDH mutations in glioma.

9.
Cancer Lett ; 495: 100-111, 2020 12 28.
Article in English | MEDLINE | ID: mdl-32768524

ABSTRACT

Early studies indicated that the testicular nuclear receptor 4 (TR4) might play key roles in altering prostate cancer (PCa) progression; however, its ability to alter PCa radiosensitivity remains unclear. Here, we found that suppressing TR4 expression promoted radiosensitivity and better suppressed PCa by modulating the protein quaking (QKI)/circZEB1/miR-141-3p/ZEB1 signaling pathway. Mechanism dissection studies revealed that TR4 could transcriptionally increase the RNA-binding protein QKI to increase circZEB1 levels, which then sponges the miR-141-3p to increase the expression of its host gene ZEB1. Preclinical studies with an in vivo mouse model further proved that combining radiation therapy (RT) with metformin promoted radiosensitivity to suppress PCa progression. Together, these results suggest that TR4 may play key roles in altering PCa radiosensitivity and show that targeting this newly identified TR4-mediated QKI/circZEB1/miR-141-3p/ZEB1 signaling pathway may help in the development of a novel RT to better suppress the progression of PCa.


Subject(s)
Metformin/administration & dosage , Prostatic Neoplasms/therapy , Radiation Tolerance/drug effects , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Up-Regulation/drug effects , Animals , Cell Line, Tumor , Humans , Male , Metformin/pharmacology , Mice , MicroRNAs/genetics , Neoplasm Transplantation , PC-3 Cells , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA, Circular/genetics , RNA-Binding Proteins/genetics , Receptors, Steroid/antagonists & inhibitors , Receptors, Thyroid Hormone/antagonists & inhibitors , Signal Transduction/radiation effects , Xenograft Model Antitumor Assays , Zinc Finger E-box-Binding Homeobox 1/genetics
10.
Oxid Med Cell Longev ; 2019: 5305014, 2019.
Article in English | MEDLINE | ID: mdl-31178964

ABSTRACT

Females develop kidney stones less frequently than males do. However, it is unclear if this gender difference is related to altered estrogen/estrogen receptor (ER) signaling. Here, we found that ER beta (ERß) signals could suppress hepatic oxalate biosynthesis via transcriptional upregulation of the glyoxylate aminotransferase (AGT1) expression. Results from multiple in vitro renal cell lines also found that ERß could function via suppressing the oxalate-induced injury through increasing the reactive oxygen species (ROS) production that led to a decrease of the renal calcium oxalate (CaOx) crystal deposition. Mechanism study results showed that ERß suppressed oxalate-induced oxidative stress via transcriptional suppression of the NADPH oxidase subunit 2 (NOX2) through direct binding to the estrogen response elements (EREs) on the NOX2 5' promoter. We further applied two in vivo mouse models with glyoxylate-induced renal CaOx crystal deposition and one rat model with 5% hydroxyl-L-proline-induced renal CaOx crystal deposition. Our data demonstrated that mice lacking ERß (ERßKO) as well as mice or rats treated with ERß antagonist PHTPP had increased renal CaOx crystal deposition with increased urinary oxalate excretion and renal ROS production. Importantly, targeting ERß-regulated NOX2 with the NADPH oxidase inhibitor, apocynin, can suppress the renal CaOx crystal deposition in the in vivo mouse model. Together, results from multiple in vitro cell lines and in vivo mouse/rat models all demonstrate that ERß may protect against renal CaOx crystal deposition via inhibiting the hepatic oxalate biosynthesis and oxidative stress-induced renal injury.


Subject(s)
Calcium Oxalate/metabolism , Estrogen Receptor beta/metabolism , Kidney Calculi/metabolism , Kidney/metabolism , Liver/metabolism , Oxidative Stress/physiology , Animals , Female , HEK293 Cells , Hep G2 Cells , Humans , Kidney/pathology , Kidney Calculi/pathology , Kidney Calculi/prevention & control , Liver/pathology , Male , Mice , Mice, Knockout
11.
EBioMedicine ; 40: 504-516, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30692044

ABSTRACT

BACKGROUND: While androgen deprivation therapy (ADT) and radiotherapy (RT) are currently used together to treat locally advanced prostate cancer (PCa), RT might have the adverse effect of increasing the PCa androgen receptor (AR) protein expression, which might then increase the resistance to continued RT. METHODS: We used multiple assays for RT sensitivity, protein and RNA expression of AR and related DDR genes, ROS level, DNA damage/repair level, cell cycle and apoptosis. All statistical comparisons were analyzed with t-test or one-way ANOVA. FINDINGS: We demonstrated that RT induced AR expression in C4-2 and CWR22Rv-1 cells. We found that combining RT and ASC-J9®, but not the antiandrogen, Enzalutamide, could increase radiosensitivity via inducing DNA damage, altering the AR mediated and DNA repair pathways, and activating apoptosis. ASC-J9® had little effects on normal bladder cells. INTERPRETATION: Targeting ionizing radiation (IR)-increased AR with the AR degradation enhancer, ASC-J9®, could increase the radiosensitivity while sparing adjacent normal tissue. Mechanism dissection revealed that ASC-J9®, but not Enzalutamide, treatment could increase radiosensitivity via inducing DNA damage, altering DNA repair pathways, as well as activating the IR-induced apoptosis via suppressing the pATR-CHK1 signals. Importantly, results from preclinical studies using an in vivo mouse model also demonstrated that combining RT with ASC-J9® to target AR led to better therapeutic efficacy to suppress PCa progression.


Subject(s)
Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Receptors, Androgen/metabolism , Animals , Cell Line, Tumor , Cell Movement/drug effects , Curcumin/analogs & derivatives , Curcumin/pharmacology , Disease Models, Animal , Disease Progression , Drug Evaluation, Preclinical , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Prostatic Neoplasms/therapy , Proteolysis , Radiation , Radiotherapy , Receptors, Androgen/genetics , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
12.
Cancer Lett ; 417: 182-191, 2018 03 28.
Article in English | MEDLINE | ID: mdl-29203251

ABSTRACT

ASC-J9® is a recently-developed androgen receptor (AR)-degradation enhancer that effectively suppresses castration resistant prostate cancer (PCa) cell proliferation and invasion. The optimal half maximum inhibitory concentrations (IC50) of ASC-J9® at various PCa cell confluences (20%, 50%, and 100%) were assessed via both short-term MTT growth assays and long-term clonogenic proliferation assays. Our results indicate that the IC50 values for ASC-J9® increased with increasing cell confluency. The IC50 values were significantly decreased in PCa AR-positive cells compared to PCa AR-negative cells or in normal prostate cells. This suggests that ASC-J9® may function mainly via targeting the AR-positive PCa cells with limited unwanted side-effects to suppress the surrounding normal prostate cells. Mechanism dissection indicated that ASC-J9® might function via altering the apoptosis signals to suppress the PCa AR-negative PC-3 cells. Preclinical studies using multiple in vitro PCa cell lines and an in vivo mouse model with xenografted castration-resistant PCa CWR22Rv1 cells demonstrated that ASC-J9® has similar AR degradation effects when dissolved in FDA-approved solvents, including DMSO, PEG-400:Tween-80 (95:5), DMA:Labrasol:Tween-80 (10:45:45), and DMA:Labrasol:Tween-20 (10:45:45). Together, results from preclinical studies suggest a potential new therapy with AR-degradation enhancer ASC-J9® may potentially be ready to be used in human clinical trials in order to better suppress PCa at later castration resistant stages.


Subject(s)
Cell Proliferation/drug effects , Curcumin/analogs & derivatives , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/metabolism , Xenograft Model Antitumor Assays , Animals , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Curcumin/pharmacology , Drug Approval , HEK293 Cells , Humans , Male , Mice, Nude , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Proteolysis/drug effects , Solutions , United States , United States Food and Drug Administration
13.
Cancer Lett ; 398: 62-69, 2017 07 10.
Article in English | MEDLINE | ID: mdl-28373004

ABSTRACT

Despite the success of androgen-deprivation therapy (ADT) with the newly developed anti-androgen enzalutamide (Enz, also known as MDV3100) to suppress castration resistant prostate cancer (CRPC) in extending patient survival by an extra 4.8 months, eventually patients die with the development of Enz resistance that may involve the induction of the androgen receptor (AR) splicing variant ARv7. Here we identify an unrecognized role of Natural Killer (NK) cells in the prostate tumor microenvironment that can be better recruited to the CRPC cells to suppress ARv7 expression resulting in suppressing the Enz resistant CRPC cell growth and invasion. Mechanism dissection revealed that CRPC cells, compared to normal prostate epithelial cells, could recruit more NK cells that might then lead to alterations of the microRNA-34 and microRNA-449 to suppress both ARv7 expression and ARv7-induced EZH2 expression to suppress CRPC cell invasion. Together, these results identify a new potential therapy using recruited NK cells to better suppress the Enz resistance and cell invasion in CRPC at the later enzalutamide resistant stage.


Subject(s)
Androgen Antagonists/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Killer Cells, Natural/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/drug effects , Alternative Splicing , Animals , Benzamides , Cell Line, Tumor , Chemotaxis, Leukocyte , Coculture Techniques , Down-Regulation , Enhancer of Zeste Homolog 2 Protein/metabolism , Humans , Killer Cells, Natural/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Invasiveness , Nitriles , Phenotype , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms, Castration-Resistant/immunology , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Isoforms , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Time Factors , Tumor Microenvironment , Xenograft Model Antitumor Assays
14.
Oncotarget ; 7(41): 66769-66775, 2016 Oct 11.
Article in English | MEDLINE | ID: mdl-27564257

ABSTRACT

Prostatitis is a common disease contributing to 8% of all urologist visits. Yet the etiology and effective treatment remain to be further elucidated. Using a non-obese diabetes mouse model that can be induced by autoimmune response for the spontaneous development of prostatitis, we found that injection of the ASC-J9® at 75 mg/Kg body weight/48 hours led to significantly suppressed prostatitis that was accompanied with reduction of lymphocyte infiltration with reduced CD4+ T cells in prostate. In vitro studies with a co-culture system also confirmed that ASC-J9® treatment could suppress the CD4+ T cell migration to prostate stromal cells. Mechanisms dissection indicated that ASC-J9® can suppress CD4+ T cell migration via decreasing the cytokine CCL2 in vitro and in vivo, and restoring CCL2 could interrupt the ASC-J9® suppressed CD4+ T cell migration. Together, results from in vivo and in vitro studies suggest that ASC-J9® can suppress prostatitis by altering the autoimmune response induced by CD4+ T cell recruitment, and using ASC-J9® may help us to develop a potential new therapy to battle the prostatitis with little side effects.


Subject(s)
Chemokine CCL2/metabolism , Curcumin/analogs & derivatives , Prostatitis/prevention & control , Signal Transduction/drug effects , Animals , Autoimmune Diseases/metabolism , Autoimmune Diseases/prevention & control , CD4-Positive T-Lymphocytes/drug effects , Cell Line , Cell Movement/drug effects , Curcumin/pharmacology , Humans , Male , Mice, Inbred NOD , Prostate/drug effects , Prostate/metabolism , Prostate/pathology , Prostatitis/metabolism , Stromal Cells/drug effects , Stromal Cells/metabolism
15.
Mol Cancer ; 15: 7, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26790618

ABSTRACT

BACKGROUND: Cancer associated fibroblasts (CAF) play important roles in tumor growth that involves inflammation and epithelial cell differentiation. Early studies suggested that estrogen receptor alpha (ERα) was expressed in stromal cells in normal prostates and prostate cancer (PCa), but the detailed functions of stromal ERα in the PCa remain to be further elucidated. METHODS: Migration and invasion assays demonstrated the presence of high levels of ERα in CAF cells (CAF.ERα(+)) suppressed PCa invasion via influencing the infiltration of tumor associated macrophages. ERα decreased CAF CCL5 secretion via suppressing the CCL5 promoter activity was examined by luciferase assay. ERα decreased CCL5 and IL-6 expression in conditioned media that was collected from CAF cell only or CAF cell co-cultured with macrophages as measured by ELISA assay. RESULTS: Both in vitro and in vivo studies demonstrated CAF.ERα(+) led to a reduced macrophage migration toward PCa via inhibiting CAF cells secreted chemokine CCL5. This CAF.ERα(+) suppressed macrophage infiltration affected the neighboring PCa cells invasion and the reduced invasiveness of PCa cells are at least partly due to reduced IL6 expression in the macrophages and CAF. CONCLUSION: Our data suggest that CAF ERα could be applied as a prognostic marker to predict cancer progression, and targeting CCL5 and IL6 may be applied as an alternative therapeutic approach to reduce M2 type macrophages and PCa invasion in PCa patients with low or little ERα expression in CAF cells.


Subject(s)
Chemokine CCL5/metabolism , Estrogen Receptor alpha/metabolism , Fibroblasts/metabolism , Interleukin-6/metabolism , Macrophages/metabolism , Prostatic Neoplasms/pathology , Tumor Microenvironment , Animals , Cell Line, Tumor , Cell Movement/drug effects , Coculture Techniques , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Macrophages/drug effects , Male , Mice, Nude , Models, Biological , Neoplasm Invasiveness , Prostatic Neoplasms/metabolism , Tumor Microenvironment/drug effects
16.
PLoS One ; 9(2): e89396, 2014.
Article in English | MEDLINE | ID: mdl-24586748

ABSTRACT

PiggyBac is a prevalent transposon system used to deliver transgenes and functionally explore the mammalian untouched genomic territory. The important features of piggyBac transposon are the relatively low insertion site preference and the ability of seamless removal from genome, which allow its potential uses in functional genomics and regenerative medicine. Efforts to increase its transposition efficiency in mammals were made through engineering the corresponding transposase (PBase) codon usage to enhance its expression level and through screening for mutant PBase variants with increased enzyme activity. To improve the safety for its potential use in regenerative medicine applications, site-specific transposition was achieved by using engineered zinc finger- and Gal4-fused PBases. An excision-prone PBase variant has also been successfully developed. Here we describe the construction of a nucleolus-predominant PBase, NP-mPB, by adding a nucleolus-predominant (NP) signal peptide from HIV-1 TAT protein to a mammalian codon-optimized PBase (mPB). Although there is a predominant fraction of the NP-mPB-tGFP fusion proteins concentrated in the nucleoli, an insertion site preference toward nucleolar organizer regions is not detected. Instead a 3-4 fold increase in piggyBac transposition efficiency is reproducibly observed in mouse and human cells.


Subject(s)
Cell Nucleolus/genetics , Cell Nucleolus/metabolism , DNA Transposable Elements/genetics , Mammals/metabolism , Transgenes/genetics , Transposases/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Codon/genetics , Genome/genetics , HEK293 Cells , HIV-1/genetics , HIV-1/metabolism , HeLa Cells , Humans , Mice , Mutagenesis, Insertional/methods , Zinc Fingers/genetics
17.
PLoS One ; 7(9): e46171, 2012.
Article in English | MEDLINE | ID: mdl-23049968

ABSTRACT

Green fluorescent protein (GFP) and its derivatives are the most widely used molecular reporters for live cell imagining. The development of organelle-specific fusion fluorescent proteins improves the labeling resolution to a higher level. Here we generate a R26 dual fluorescent protein reporter mouse, activated by Cre-mediated DNA recombination, labeling target cells with a chromatin-specific enhanced green fluorescence protein (EGFP) and a plasma membrane-anchored monomeric cherry fluorescent protein (mCherry). This dual labeling allows the visualization of mitotic events, cell shapes and intracellular vesicle behaviors. We expect this reporter mouse to have a wide application in developmental biology studies, transplantation experiments as well as cancer/stem cell lineage tracing.


Subject(s)
Green Fluorescent Proteins/metabolism , Integrases/metabolism , Alleles , Animals , Cells, Cultured , Green Fluorescent Proteins/genetics , Integrases/genetics , Mice , Mice, Transgenic , Mitosis/genetics , Mitosis/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...