Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Neurol Res ; : 1-11, 2024 May 12.
Article in English | MEDLINE | ID: mdl-38735062

ABSTRACT

OBJECTIVE: Cerebral ischemia is a neurological disorder that leads to permanent disability. This research focuses on exploring the ameliorative effects of lipid nanoparticle (LNP)-encapsulated lncRNA DLX6-AS1 knockdown in cerebral ischemic injury via the Nrf2/HO-1/NLRP3 axis. METHODS: LNP-encapsulated lncRNA DLX6-AS1 was prepared. Cerebral ischemic injury mouse models were established utilizing middle cerebral artery occlusion (MCAO). The mice were treated by intravenous injection of LNP-encapsulated lncRNA DLX6-AS1. The neurological deficits, Inflammatory factor levels, pathological characteristics were observed. In vitro N2a cell oxygen and glucose deprivation (OGD) models were established, and the cells were treated with LNP-encapsulated lncRNA DLX6-AS1 or Nrf2 inhibitor (ML385). Cell viability and apoptosis were tested. DLX6-AS1, Nrf2, HO-1, and NLRP3 expression levels were assessed. RESULTS: LncRNA DLX6-AS1 levels were elevated in the brain tissues of mice with cerebral ischemic injury and OGD-induced N2a cells. LNP-encapsulated DLX6-AS1 siRNA (si-DLX6-AS1) improved neurological deficit scores, reduced the levels of inflammatory factors, improved brain tissue pathological damage, and raised the number of survival neurons in CA1. LNP-encapsulated si-DLX6-AS1 ameliorated the OGD-induced N2a cell viability decrease and apoptosis rate increase, and ML385 (Nrf2 inhibitor) reversed the ameliorative effects of LNP-encapsulated si-DLX6-AS1. In cerebral ischemic injury mice and OGD-induced N2a cells, Nrf2 and HO-1 levels were reduced and NLRP3 levels were increased. LNP-encapsulated si-DLX6-AS1 raised Nrf2 and HO-1 levels and reduced NLRP3 levels. Nrf2 inhibitor ML385 treatment reversed the ameliorative effects of LNP-encapsulated si-DLX6-AS1 on OGD-induced N2a cell viability and apoptosis. CONCLUSION: Lipid nanoparticle-encapsulated si-DLX6-AS1 ameliorates cerebral ischemic injury via the Nrf2/HO-1/NLRP3 axis.

2.
Environ Sci Pollut Res Int ; 30(16): 46383-46393, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36719573

ABSTRACT

Bisphenol A (BPA) is considered a contaminant of emerging concern and interferes with the normal activities of living organisms. The toxicity of BPA is evident in animals and terrestrial plants. However, the response of aquatic plants to low BPA concentrations is still unclear. In the present study, effects of varying BPA loadings (targeting at 0.01, 0.1, and 1 mg/L) on the growth and reproductive traits of the dioecious annual submerged macrophyte Vallisneria natans were assessed through a 5-month experiment. The results showed that BPA inhibited the elongation of V. natans leaves but resulted in an increase in leaf number and ramet number under the highest BPA loading treatment (targeting at 1 mg/L). In addition, detectable biochemical changes in the total carbon and soluble sugar contents were found, which both were significantly higher at the highest BPA loading treatment. However, the total biomass did not alter significantly after the BPA treatments, indicating that BPA did not induce direct toxic effects on the growth of V. natans. At the highest BPA loading treatment, female individuals of V. natans allocated less number for ramet than male ones, showing a clear sexual dimorphism. No significant differences between the five treatments were found for the flower or fruit traits, while the germination rate was significantly inhibited for the seeds collected from the highest BPA loading treatment. In conclusion, V. natans tolerated low concentrations of BPA by making a trade-off between ramet (leaf) number and leaf elongation, as well as modulating the total carbon and soluble sugar contents. However, serious consequence of decline in seed viability implied that the impact of BPA on plant reproduction were usually underestimated.


Subject(s)
Hydrocharitaceae , Plants , Animals , Biomass , Seeds , Hydrocharitaceae/physiology
3.
Sci Adv ; 8(17): eabh3635, 2022 04 29.
Article in English | MEDLINE | ID: mdl-35476451

ABSTRACT

MYC regulates multiple gene programs, raising questions about the potential selectivity and downstream transcriptional consequences of MYC inhibitors as cancer therapeutics. Here, we examined the effect of a small-molecule MYC inhibitor, MYCi975, on the MYC/MAX cistromes, epigenome, transcriptome, and tumorigenesis. Integrating these data revealed three major classes of MYCi975-modulated gene targets: type 1 (down-regulated), type 2 (up-regulated), and type 3 (unaltered). While cell cycle and signal transduction pathways were heavily targeted by MYCi, RNA biogenesis and core transcriptional pathway genes were spared. MYCi975 altered chromatin binding of MYC and the MYC network family proteins, and chromatin accessibility and H3K27 acetylation alterations revealed MYCi975 suppression of MYC-regulated lineage factors AR/ARv7, FOXA1, and FOXM1. Consequently, MYCi975 synergistically sensitized resistant prostate cancer cells to enzalutamide and estrogen receptor-positive breast cancer cells to 4-hydroxytamoxifen. Our results demonstrate that MYCi975 selectively inhibits MYC target gene expression and provide a mechanistic rationale for potential combination therapies.


Subject(s)
Breast Neoplasms , Epigenomics , Chromatin/genetics , Gene Expression , Humans , Male , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism
4.
Cancer Res ; 82(11): 2110-2123, 2022 06 06.
Article in English | MEDLINE | ID: mdl-35405009

ABSTRACT

Androgen receptor (AR) pathway inhibitors are the mainstay treatment for advanced prostate cancer, but resistance to therapy is common. Here, we used a CRISPR activation screen in metastatic castration-sensitive prostate cancer cells to identify genes that promote resistance to AR inhibitors. Activation of the TGFß target gene paired-related homeobox2 (PRRX2) promoted enzalutamide resistance. PRRX2 expression was the highest in double-negative prostate cancer (DNPC), which lack AR signaling and neuroendocrine differentiation, and a PRRX2-related gene signature identified a subset of patients with DNPC with reduced overall survival. PRRX2-expressing cells showed alterations in the CDK4/6/Rb/E2F and BCL2 pathways. Accordingly, treatment with CDK4/6 and BCL2 inhibitors sensitized PRRX2-expressing, castration-resistant tumors to enzalutamide. Overall, PRRX2 was identified as a driver of enzalutamide resistance. The PRRX2 signature merits investigation as a biomarker of enzalutamide resistance in prostate cancer that could be reversed with CDK4/6 and BCL2 inhibitors. SIGNIFICANCE: PRRX2 mediates enzalutamide resistance via activation of the E2F and BCL2 pathways, which can be targeted with CDK4/6 and BCL2 inhibitors to reverse resistance.


Subject(s)
Antineoplastic Agents , Prostatic Neoplasms, Castration-Resistant , Androgen Receptor Antagonists/pharmacology , Antineoplastic Agents/pharmacology , Benzamides , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats , Drug Resistance, Neoplasm/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Nitriles/therapeutic use , Phenylthiohydantoin , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Androgen/metabolism
5.
Prostate Cancer Prostatic Dis ; 25(3): 463-471, 2022 09.
Article in English | MEDLINE | ID: mdl-34035460

ABSTRACT

BACKGROUND: Inflammatory bowel disease (IBD) has been implicated as a risk factor for prostate cancer, however, the mechanism of how IBD leads to prostate tumorigenesis is not known. Here, we investigated whether chronic intestinal inflammation leads to pro-inflammatory changes associated with tumorigenesis in the prostate. METHODS: Using clinical samples of men with IBD who underwent prostatectomy, we analyzed whether prostate tumors had differences in lymphocyte infiltrate compared to non-IBD controls. In a mouse model of chemically-induced intestinal inflammation, we investigated whether chronic intestinal inflammation could be transferred to the wild-type mouse prostate. In addition, mouse prostates were evaluated for activation of pro-oncogenic signaling and genomic instability. RESULTS: A higher proportion of men with IBD had T and B lymphocyte infiltration within prostate tumors. Mice with chronic colitis showed significant increases in prostatic CD45 + leukocyte infiltration and elevation of three pro-inflammatory cytokines-TIMP-1, CCL5, and CXCL1 and activation of AKT and NF-kB signaling pathways. Lastly, mice with chronic colitis had greater prostatic oxidative stress/DNA damage, and prostate epithelial cells had undergone cell cycle arrest. CONCLUSIONS: These data suggest chronic intestinal inflammation is associated with an inflammatory-rich, pro-tumorigenic prostatic phenotype which may explain how gut inflammation fosters prostate cancer development in men with IBD.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Prostatic Neoplasms , Animals , Carcinogenesis , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Dextran Sulfate/adverse effects , Disease Models, Animal , Humans , Inflammation , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/genetics , Male , Mice , Mice, Inbred C57BL , Prostate/pathology , Prostatic Neoplasms/genetics
7.
J Asian Nat Prod Res ; 24(8): 754-760, 2022 Aug.
Article in English | MEDLINE | ID: mdl-34647847

ABSTRACT

Quercetin (1) was converted into quercetin 7-O-succinyl glucoside (2) by used Bacillus amyloliquefaciens FJ18 as a solvent-resistant whole-cell biocatalyst. The structure of the new compound was confirmed by LC-MS analysis and NMR spectroscopy. The water-solubility of this novel quercetin 7-O-succinyl glucoside (2) was approximately 1000 times higher than that of native quercetin (2). Quercetin (1) and quercetin 7-O-succinyl glucoside (2) exhibited significant DPPH scavenging capacity with IC50 values of 23.55 and 36.05 µM, respectively. Both compounds showed moderate cytotoxic effects against the two human cancer cell lines (MCF-7 and HepG2) with IC50 values ranging from 39.45-63.38 µM.


Subject(s)
Antioxidants , Quercetin , Antioxidants/pharmacology , Glucosides/chemistry , Humans , Molecular Structure , Rutin , Water
8.
Inflamm Res ; 71(1): 93-106, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34767031

ABSTRACT

INTRODUCTION: Cerebral ischemic injury is associated with long-term disability. Dexmedetomidine (Dex) can exert neuroprotective effects on cerebral ischemic/reperfusion injury. The present study explored the mechanism of Dex in cerebral ischemic injury. MATERIALS AND METHODS: To this end, the permanent middle cerebral artery occlusion (p-MCAO) mouse model was established and treated with Dex or/and Nrf2 inhibitor ML385. Subsequently, microglia were subjected to oxygen-glucose deprivation (OGD) in sugar-free environment and thereafter treated with Dex, Nrf2 inhibitor, and NLRP3 lentiviral overexpression vector, respectively. RESULTS: Dex alleviated the neurobehavioral deficit of p-MCAO mice, reduced brain water content, relieved pathological changes, and reduced cerebral infarction size. Dex promoted the polarization of microglia from M1 to M2, thus ameliorating oxidative stress and inflammatory responses. Our results showed that Dex promoted M2-polarization of microglia in vivo and in vitro by promoting HO-1 expression via Nrf2 nuclear import. Moreover, the Nrf2/HO-1 axis inhibited the activation of NLRP2 inflammasome and NLRP3 overexpression reversed the effect of Dex. CONCLUSION: In conclusion, Dex promoted M2-polarization of microglia and attenuated oxidative stress and inflammation, and thus protected against cerebral ischemic injury by activating the Nrf2/HO-1 pathway and inhibiting NLRP3 inflammasome.


Subject(s)
Brain Ischemia , Dexmedetomidine , Neuroprotective Agents , Reperfusion Injury , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis Regulatory Proteins , Brain Ischemia/drug therapy , Brain Ischemia/prevention & control , Dexmedetomidine/pharmacology , Dexmedetomidine/therapeutic use , Heme Oxygenase-1 , Membrane Proteins , Mice , Microglia/metabolism , NF-E2-Related Factor 2/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Reperfusion Injury/metabolism , Signal Transduction
9.
Brain Res Bull ; 175: 81-89, 2021 10.
Article in English | MEDLINE | ID: mdl-34265390

ABSTRACT

OBJECTIVE: Long non-coding RNAs (lncRNAs) have diagnostic and therapeutic values in the setting of ischemic stroke (IS). Here, we evaluated the value of myocardial infarction-associated transcript (MIAT) in IS with the involvement of microRNA (miR)-874-3p/interleukin (IL) 1B. METHODS: MIAT, miR-874-3p and IL1B levels in serum of patients with IS were measured. A middle cerebral artery occlusion (MCAO) model was established in mice. MCAO mice were injected with Agomir of miR-874-3p, shRNA or overexpression vector of MIAT or siRNA of IL1B. Subsequently, behavioral activities and neurological function of mice were assessed. The number of Nissl bodies, brain damage, neuronal apoptosis and inflammatory factors in brain tissues of mice were measured. The targeting relationship between MIAT and miR-874-3p, as well as that between miR-874-3p and IL1B was explored. RESULTS: In patients with IS, MIAT and IL1B were up-regulated and miR-874-3p was down-regulated. MIAT absorbed miR-874-3p while miR-874-3p targeted IL1B. Silencing of MIAT or IL1B, or promotion of miR-874-3p improved behavioral activities and neurological function of mice, reduced the number of Nissl bodies, as well as improved brain damage, neuronal apoptosis and inflammation. Overexpression of miR-874-3p abrogated up-regulated MIAT-mediated influence on MCAO mice. CONCLUSION: Shortly, this study figures out that MIAT impairs neurological function in IS via up-regulating miR-874-3p-targeted IL1B.


Subject(s)
Interleukin-1beta/genetics , Ischemic Stroke/genetics , Ischemic Stroke/physiopathology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Animals , Behavior, Animal , Brain/pathology , Female , Gene Silencing , Gene Targeting , Humans , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Interleukin-1beta/blood , Ischemic Stroke/pathology , Male , Mice , Middle Aged , Up-Regulation
10.
Cancer Res ; 81(8): 2157-2170, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33637566

ABSTRACT

Neuroendocrine prostate cancer (NEPC) is an aggressive subtype of prostate cancer with poor prognosis, and there is a critical need for novel therapeutic approaches. NEPC is associated with molecular perturbation of several pathways, including amplification of MYCN. Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase involved in the pathogenesis of neuroblastoma and other malignancies where it cooperates with N-Myc. We previously identified the first case of ALK F1174C-activating mutation in a patient with de novo NEPC who responded to the ALK inhibitor, alectinib. Here, we show that coactivation of ALK and N-Myc (ALK F1174C/N-Myc) is sufficient to transform mouse prostate basal stem cells into aggressive prostate cancer with neuroendocrine differentiation in a tissue recombination model. A novel gene signature from the ALK F1174C/N-Myc tumors was associated with poor outcome in multiple human prostate cancer datasets. ALK F1174C and ALK F1174C/N-Myc tumors displayed activation of the Wnt/ß-catenin signaling pathway. Chemical and genetic ALK inhibition suppressed Wnt/ß-catenin signaling and tumor growth in vitro in NEPC and neuroblastoma cells. ALK inhibition cooperated with Wnt inhibition to suppress NEPC and neuroblastoma proliferation in vitro and tumor growth and metastasis in vivo. These findings point to a role for ALK signaling in NEPC and the potential of cotargeting the ALK and Wnt/ß-catenin pathways in ALK-driven tumors. Activated ALK and N-Myc are well known drivers in neuroblastoma development, suggesting potential similarities and opportunities to elucidate mechanisms and therapeutic targets in NEPC and vice versa. SIGNIFICANCE: These findings demonstrate that coactivation of ALK and N-Myc induces NEPC by stimulating the Wnt/ß-catenin pathway, which can be targeted therapeutically.


Subject(s)
Anaplastic Lymphoma Kinase/metabolism , Carcinoma, Neuroendocrine/etiology , N-Myc Proto-Oncogene Protein/metabolism , Prostatic Neoplasms/etiology , Wnt Signaling Pathway , Anaplastic Lymphoma Kinase/antagonists & inhibitors , Anaplastic Lymphoma Kinase/genetics , Animals , Carbazoles/therapeutic use , Carcinoma, Neuroendocrine/drug therapy , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/pathology , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Humans , Male , Mice , Mutation , N-Myc Proto-Oncogene Protein/genetics , Neoplastic Stem Cells , Neuroblastoma/drug therapy , Neuroblastoma/etiology , Neuroblastoma/pathology , Piperidines/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/therapeutic use , Exome Sequencing , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics
11.
Cancer Res ; 81(2): 248-253, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33087323

ABSTRACT

MYC is a highly validated oncogenic transcription factor and cancer target. However, the disordered nature of this protein has made it a challenging target, with no clinical stage, direct small-molecule MYC inhibitors available. Recent work leveraging a large in silico chemical library and a rapid in vivo screen has expanded the chemotypes of direct small-molecule inhibitors (MYCi). Novel MYCi represent a class of improved MYC chemical probes that bind directly to MYC to inhibit its function and to promote its degradation by enhancing GSK3ß-mediated phosphorylation. One of these compounds, MYCi975, has shown remarkable tolerability and efficacy in vivo and is associated with a selective effect on MYC target gene expression. Additional effects of MYCi on the tumor immune microenvironment including immune cell infiltration and upregulation of PD-L1 expression provide a rationale for combining MYCi with anti-PD-1/PD-L1 therapy to enhance antitumor efficacy. Our strategy for developing MYCi demonstrates an efficient way to identify selective and well-tolerated MYC inhibitors. The new MYCi provide tools for probing MYC function and serve as starting points for the development of novel anti-MYC therapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Neoplasms/drug therapy , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Tumor Microenvironment/drug effects , Animals , Humans , Neoplasms/metabolism , Neoplasms/pathology , Proto-Oncogene Proteins c-myc/genetics
12.
Blood ; 137(12): 1603-1614, 2021 03 25.
Article in English | MEDLINE | ID: mdl-32967010

ABSTRACT

The initiation and progression of diffuse large B-cell lymphoma (DLBCL) is governed by genetic and epigenetic aberrations. As the most abundant eukaryotic messenger RNA (mRNA) modification, N6-methyladenosine (m6A) is known to influence various fundamental bioprocesses by regulating the target gene; however, the function of m6A modifications in DLBCL is unclear. PIWI-interacting RNAs (piRNAs) have been indicated to be epigenetic effectors in cancer. Here, we show that high expression of piRNA-30473 supports the aggressive phenotype of DLBCL, and piRNA-30473 depletion decreases proliferation and induces cell cycle arrest in DLBCL cells. In xenograft DLBCL models, piRNA-30473 inhibition reduces tumor growth. Moreover, piRNA-30473 is significantly associated with overall survival in a univariate analysis and is statistically significant after adjusting for the National Comprehensive Cancer Network-International Prognostic Index in the multivariate analysis. Additional studies demonstrate that piRNA-30473 exerts its oncogenic role through a mechanism involving the upregulation of WTAP, an m6A mRNA methylase, and thus enhances the global m6A level. Integrating transcriptome and m6A-sequencing analyses reveals that WTAP increases the expression of its critical target gene, hexokinase 2 (HK2), by enhancing the HK2 m6A level, thereby promoting the progression of DLBCL. Together, the piRNA-30473/WTAP/HK2 axis contributes to tumorigenesis by regulating m6A RNA methylation in DLBCL. Furthermore, by comprehensively analyzing our clinical data and data sets, we discover that the m6A regulatory genes piRNA-30473 and WTAP improve survival prediction in DLBCL patients. Our study highlights the functional importance of the m6A modification in DLBCL and might assist in the development of a prognostic stratification and therapeutic approach for DLBCL.


Subject(s)
Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Lymphoma, Large B-Cell, Diffuse/genetics , RNA, Small Interfering/genetics , Epigenesis, Genetic , Humans , Lymphoma, Large B-Cell, Diffuse/diagnosis , Methyltransferases/genetics , Prognosis , RNA, Messenger/genetics
13.
Cancer Cell ; 36(5): 483-497.e15, 2019 11 11.
Article in English | MEDLINE | ID: mdl-31679823

ABSTRACT

Small molecules that directly target MYC and are also well tolerated in vivo will provide invaluable chemical probes and potential anti-cancer therapeutic agents. We developed a series of small-molecule MYC inhibitors that engage MYC inside cells, disrupt MYC/MAX dimers, and impair MYC-driven gene expression. The compounds enhance MYC phosphorylation on threonine-58, consequently increasing proteasome-mediated MYC degradation. The initial lead, MYC inhibitor 361 (MYCi361), suppressed in vivo tumor growth in mice, increased tumor immune cell infiltration, upregulated PD-L1 on tumors, and sensitized tumors to anti-PD1 immunotherapy. However, 361 demonstrated a narrow therapeutic index. An improved analog, MYCi975 showed better tolerability. These findings suggest the potential of small-molecule MYC inhibitors as chemical probes and possible anti-cancer therapeutic agents.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , B7-H1 Antigen/pharmacology , Drug Discovery/methods , Neoplasms/drug therapy , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , B7-H1 Antigen/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Feasibility Studies , Female , Humans , Male , Mice , Neoplasms/immunology , Neoplasms/pathology , Phosphorylation/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Proto-Oncogene Proteins c-myc/metabolism , Threonine/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
14.
Cell Death Dis ; 10(11): 801, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31641103

ABSTRACT

The EPHB4 receptor is implicated in the development of several epithelial tumors and is a promising therapeutic target, including in prostate tumors in which EPHB4 is overexpressed and promotes tumorigenicity. Here, we show that high expression of EPHB4 correlated with poor survival in prostate cancer patients and EPHB4 inhibition induced cell death in both hormone sensitive and castration-resistant prostate cancer cells. EPHB4 inhibition reduced expression of the glucose transporter, GLUT3, impaired glucose uptake, and reduced cellular ATP levels. This was associated with the activation of endoplasmic reticulum stress and tumor cell death with features of immunogenic cell death (ICD), including phosphorylation of eIF2α, increased cell surface calreticulin levels, and release of HMGB1 and ATP. The changes in tumor cell metabolism after EPHB4 inhibition were associated with MYC downregulation, likely mediated by the SRC/p38 MAPK/4EBP1 signaling cascade, known to impair cap-dependent translation. Together, our study indicates a role for EPHB4 inhibition in the induction of immunogenic cell death with implication for prostate cancer therapy.


Subject(s)
Endoplasmic Reticulum Stress/immunology , Immunogenic Cell Death/immunology , Prostatic Neoplasms/immunology , Receptor, EphB4/antagonists & inhibitors , Animals , Cell Line, Tumor , Humans , Male , Mice , PC-3 Cells , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptor, EphB4/genetics , Receptor, EphB4/immunology , Receptor, EphB4/metabolism , Signal Transduction
15.
Clin Cancer Res ; 25(6): 1923-1935, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30610101

ABSTRACT

PURPOSE: How exosomal RNAs released within the bone marrow microenvironment affect proteasome inhibitors' (PI) sensitivity of multiple myeloma is currently unknown. This study aims to evaluate which exosomal RNAs are involved and by which molecular mechanisms they exert this function.Experimental Design: Exosomes were characterized by dynamic light scattering, transmission electron microscopy, and Western blot analysis. Coculture experiments were performed to assess exosomal RNAs transferring from mesenchymal stem cells (MSC) to multiple myeloma cells. The role of PSMA3-AS1 in PI sensitivity was further evaluated in vivo. To determine the prognostic significance of circulating exosomal PSMA3 and PSMA3-AS1, a cohort of patients with newly diagnosed multiple myeloma was enrolled to study. Cox regression models and Kaplan-Meier curves were used to analyze progression-free survival (PFS) and overall survival (OS). RESULTS: We identified that PSMA3 and PSMA3-AS1 in MSCs could be packaged into exosomes and transferred to myeloma cells, thus promoting PI resistance. PSMA3-AS1 could form an RNA duplex with pre-PSMA3, which transcriptionally promoted PSMA3 expression by increasing its stability. In xenograft models, intravenously administered siPSMA3-AS1 was found to be effective in increasing carfilzomib sensitivity. Moreover, plasma circulating exosomal PSMA3 and PSMA3-AS1 derived from patients with multiple myeloma were significantly associated with PFS and OS. CONCLUSIONS: This study suggested a unique role of exosomal PSMA3 and PSMA3-AS1 in transmitting PI resistance from MSCs to multiple myeloma cells, through a novel exosomal PSMA3-AS1/PSMA3 signaling pathway. Exosomal PSMA3 and PSMA3-AS1 might act as promising therapeutic targets for PI resistance and prognostic predictors for clinical response.


Subject(s)
Drug Resistance, Neoplasm/genetics , Multiple Myeloma/drug therapy , Proteasome Inhibitors/pharmacology , RNA, Antisense/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Cell Proliferation , Exosomes/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mesenchymal Stem Cells/metabolism , Mice , Middle Aged , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Prognosis , Progression-Free Survival , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/therapeutic use , Protein Stability , Signal Transduction/genetics , Xenograft Model Antitumor Assays
16.
RSC Adv ; 9(51): 29652-29658, 2019 Sep 18.
Article in English | MEDLINE | ID: mdl-35531511

ABSTRACT

A series of dicationic ionic liquids (ILs) including [PF6][(PYR)C4(MIM)][Cl], [PF6][(PYR)C4(PYR)][Cl], [PF6][(PYR)C5(MIM)][Cl], and [PF6][(PYR)C5(PYR)][Cl], and monocationic ILs including [(PYR)C4Cl][PF6], [(PYR)C5Cl][PF6], [(MIM)C2COOH][PF6] and [(PYR)C2COOH][PF6] were synthesized. Their thermal stability and melting points were determined. Their solubility with organic solvents and the miscibility with water were investigated. These functional ILs are hydrophilic at high temperatures and they are hydrophobic at low temperatures, which enable the effective isolation of the resulting reducing sugar. High yields of reducing sugar were obtained for corn stalk after 8 h (20.73%) and potato starch after 6 h (72.50%) by the treatment with the mixture of [PF6][(PYR)C4(PYR)][Cl] and [(PYR)C2COOH][PF6]. The reuse of dicationic and monocationic ILs was successfully performed and no significant reduction in yields of reducing sugar was observed. These functional ILs have important implications in the design of homogeneous and heterogeneous systems with water and organic solvents, which could be used to satisfy some specific applications.

17.
Stem Cells ; 37(2): 247-256, 2019 02.
Article in English | MEDLINE | ID: mdl-30353595

ABSTRACT

The characteristics of mesenchymal stromal cells (MSCs) which derived from multiple myeloma (MM) patients are typically impaired in osteogenic differentiation. However, the underlying molecular mechanisms need to be further investigated. lncRNAs are emerging as critical regulation molecules in oncogenic pathways. In this study, we identified that bioactive lncRNA HOXC-AS3, which is transcribed in opposite to HOXC10, was presented in MSCs derived from bone marrow (BM) of MM patients (MM-MSCs). HOXC-AS3 was able to interact with HOXC10 at the overlapping parts and this interaction increased HOXC10 stability, then promoted its expression, conferring osteogenesis repression to MM-MSCs. In mouse models, intravenously administered siHOXC-AS3 was proven to be effective in prevention of bone loss, sustained by both anticatabolic activities and bone-forming. These data showed that lncHOXC-AS3 was required for osteogenesis in BM-MSCs by enhancing HOXC10 expression. Our finding thus unveils a novel insight for the potential clinical significance of lncRNA HOXC-AS3 as a therapeutic target for bone disease in MM. Stem Cells 2019;37:247-256.


Subject(s)
Homeodomain Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Multiple Myeloma/metabolism , Oligonucleotides, Antisense/metabolism , Osteoblasts/metabolism , RNA, Long Noncoding/metabolism , Animals , Case-Control Studies , Cell Differentiation/physiology , Cell Line, Tumor , Cells, Cultured , Female , Heterografts , Homeodomain Proteins/genetics , Humans , Mesenchymal Stem Cells/cytology , Mice , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Oligonucleotides, Antisense/genetics , Osteogenesis , RNA, Long Noncoding/genetics , Transfection , Up-Regulation
18.
Mol Pharm ; 15(11): 5387-5396, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30339013

ABSTRACT

Treatment failure remains a main challenge in the management of high-risk multiple myeloma (MM) even with the expanding repertoire of new drugs. Combinatorial therapy is considered an encouraging strategy that can overcome the compensatory mechanisms and undesirable off-target effects that limit the benefits of many prospective agents. Preliminary results of a current phase I trial have indicated that the new BET bromodomain inhibitor OTX015 has favorable activity and tolerability. However, OTX015 is not efficacious enough as a monotherapy. Here, we provide evidence that synergistic drug combinations with OTX015 were generally more specific to particular cellular contexts than single agent activities. In addition, pairing OTX015 with three classes of drugs dramatically enhanced the antitumor activity in mouse models of disseminated human myeloma. Our studies further underscored that the BET inhibitor OTX015 sensitized MM cells by interrupting several pathways and genes critical for MM cell proliferation and drug response, which provided the rationale for multiple myeloma therapy with OTX015 combined with conventional chemotherapeutic drugs. Thus, the context specificity of synergistic combinations not only provide profound insights into therapeutically relevant selectivity but also improve control of complex biological systems.


Subject(s)
Acetanilides/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Multiple Myeloma/drug therapy , Acetanilides/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Drug Synergism , Heterocyclic Compounds, 3-Ring/therapeutic use , Humans , Mice , Multiple Myeloma/pathology , Prospective Studies , Proteins/antagonists & inhibitors , Xenograft Model Antitumor Assays
19.
Mol Pharm ; 15(9): 4139-4147, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30048594

ABSTRACT

Several studies demonstrate that the bromodomain inhibitor OTX015 has an antitumor activity in cancers. However, translation of these data to molecules suitable for clinical development has yet to be accomplished in multiple myeloma (MM). Here, we identified genes and biologic processes that substantiated the antimyeloma activity of OTX015 with global transcriptomics. OTX015 exerted a strong antiproliferative effect and induced cell cycle arrest in vitro. Gene expression profiling uncovered that OTX015 targeted NF-κB, EGFR, cell cycle regulation, and the cancer proliferation signaling pathway. Gene expression signatures displaying various levels of sensitivity to OTX015 were also identified. The data also showed that oral administration of OTX015 displayed significant antitumor activity in the mice model of disseminated human myeloma. In addition, our study provided the first evidence and rationale that OTX015 could promote osteoblast differentiation of mesenchymal stem cells (MSCs) and inhibited osteoclast formation and resorption in vivo experiments. Herein our results expanded the understanding of the mechanism for BET inhibitors OTX015 in MM. Our study provided an impressive basis for the clinical application of the novel antimyeloma agent OTX015 and uncovered signaling pathways that may play key roles in myeloma cell proliferation.


Subject(s)
Acetanilides/therapeutic use , Antineoplastic Agents/therapeutic use , Heterocyclic Compounds, 3-Ring/therapeutic use , Multiple Myeloma/drug therapy , Proteins/antagonists & inhibitors , Animals , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Mice , Multiple Myeloma/metabolism , Osteogenesis/drug effects , Xenograft Model Antitumor Assays
20.
Bioinformatics ; 34(21): 3609-3615, 2018 11 01.
Article in English | MEDLINE | ID: mdl-29850860

ABSTRACT

Motivation: Molecular analyses suggest that myeloma is composed of distinct sub-types that have different molecular pathologies and various response rates to certain treatments. Drug responses in multiple myeloma (MM) are usually recorded as a multi-level ordinal outcome. One of the goals of drug response studies is to predict which response category any patients belong to with high probability based on their clinical and molecular features. However, as most of genes have small effects, gene-based models may provide limited predictive accuracy. In that case, methods for predicting multi-level ordinal drug responses by incorporating biological pathways are desired but have not been developed yet. Results: We propose a pathway-structured method for predicting multi-level ordinal responses using a two-stage approach. We first develop hierarchical ordinal logistic models and an efficient quasi-Newton algorithm for jointly analyzing numerous correlated variables. Our two-stage approach first obtains the linear predictor (called the pathway score) for each pathway by fitting all predictors within each pathway using the hierarchical ordinal logistic approach, and then combines the pathway scores as new predictors to build a predictive model. We applied the proposed method to two publicly available datasets for predicting multi-level ordinal drug responses in MM using large-scale gene expression data and pathway information. Our results show that our approach not only significantly improved the predictive performance compared with the corresponding gene-based model but also allowed us to identify biologically relevant pathways. Availability and implementation: The proposed approach has been implemented in our R package BhGLM, which is freely available from the public GitHub repository https://github.com/abbyyan3/BhGLM.


Subject(s)
Biological Phenomena , Multiple Myeloma , Algorithms , Bayes Theorem , Humans , Logistic Models , Multiple Myeloma/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...