Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 135
Filter
1.
Bioconjug Chem ; 34(1): 238-247, 2023 01 18.
Article in English | MEDLINE | ID: mdl-36516871

ABSTRACT

As a counterpart to antibody-drug conjugates (ADCs), aptamer-drug conjugates (ApDCs) have been considered a promising strategy for targeted therapy due to the various benefits of aptamers. However, an aptamer merely serves as a targeting ligand in ApDCs, whereas the antibody enables the unexpected therapeutic efficacy of ADCs through antibody-dependent cellular cytotoxicity (ADCC). In this study, we developed a tumor-specific aptamer with an effector function and used it to confirm the feasibility of more potent ApDCs. First, we designed a nucleolin (NCL)-binding G-quadruplex (GQ) library based on the ability of NCL to bind to telomeric sequences. We then identified a bifunctional GQ aptamer (BGA) inhibiting the catalytic activity of topoisomerase 1 (TOP1) by forming an irreversible cleavage complex. Our BGA specifically targeted NCL-positive MCF-7 cells, exhibiting antiproliferative activity, and this suggested that tumor-specific therapeutic aptamers can be developed by using a biased library to screen aptamer candidates for functional targets. Finally, we utilized DM1, which has a synergistic interaction with TOP1 inhibitors, as a conjugated drug. BGA-DM1 exerted an anticancer effect 20-fold stronger than free DM1 and even 10-fold stronger than AS1411 (NCL aptamer)-DM1, highlighting our approach to develop synergistic ApDCs. Therefore, we anticipate that our library might be utilized for the identification of aptamers with effector functions. Furthermore, by employing such aptamers and appropriate drugs, synergistic ApDCs can be developed for targeted cancer therapy in a manner distinct from how ADCs exhibit additional therapeutic efficacy.


Subject(s)
Aptamers, Nucleotide , DNA Topoisomerases, Type I , RNA-Binding Proteins , Humans , Aptamers, Nucleotide/pharmacology , Aptamers, Nucleotide/metabolism , MCF-7 Cells , Phosphoproteins/metabolism , RNA-Binding Proteins/drug effects , RNA-Binding Proteins/metabolism , DNA Topoisomerases, Type I/drug effects , DNA Topoisomerases, Type I/metabolism , Drug Synergism , Nucleolin
2.
J Diabetes Res ; 2022: 7680513, 2022.
Article in English | MEDLINE | ID: mdl-35308095

ABSTRACT

To explore the relevant RNA-binding proteins (RBPs) and alternative splicing events (ASEs) in diabetic retinopathy (DR). We devised a comprehensive work to integrate analyses of the differentially expressed genes, including differential RBPs, and variable splicing characteristics related to DR in human retinal endothelial cells induced by low glucose and high glucose in dataset GSE117238. A total of 2320 differentially expressed genes (DEGs) were identified, including 1228 upregulated genes and 1092 downregulated genes. Further analysis screened out 232 RBP genes, and 42 AS genes overlapped DEGs. We selected high expression and consistency six RBP genes (FUS, HNRNPA2B1, CANX, EIF1, CALR, and POLR2A) for coexpression analysis. Through analysis, we found eight RASGs (MDM2, GOLGA2P7, NFE2L1, KDM4A, FAM111A, CIRBP, IDH1, and MCM7) that could be regulated by RBP. The coexpression network was conducted to further elucidate the regulatory and interaction relationship between RBPs and AS. Apoptotic progress, protein phosphorylation, and NF-kappaB cascade revealed by the functional enrichment analysis of RASGs regulated by RBPs were closely related to diabetic retinopathy. Furthermore, the expression of differentially expressed RBPs was validated by qRT-PCR in mouse retinal microvascular endothelial cells and retinas from the streptozotocin mouse model. The results showed that Fus, Hnrnpa2b1, Canx, Calr, and Polr2a were remarkedly difference in high-glucose-treated retinal microvascular endothelial cells and Fus, Hnrnpa2b1, Canx, and Calr were remarkedly difference in retinas from streptozotocin-induced diabetic mice compared to control. The regulatory network between identified RBPs and RASGs suggests the presence of several signaling pathways possibly involved in the pathogenesis of DR. The verified RBPs should be further addressed by future studies investigating associations between RBPs and the downstream of AS, as they could serve as potential biomarkers and targets for DR.


Subject(s)
Alternative Splicing/physiology , Blood Glucose/metabolism , Endothelial Cells/drug effects , RNA-Binding Proteins/metabolism , Retina/drug effects , Alternative Splicing/drug effects , Animals , Disease Models, Animal , Endothelial Cells/metabolism , Mice , Mice, Inbred NOD , RNA-Binding Proteins/drug effects , Real-Time Polymerase Chain Reaction/methods , Real-Time Polymerase Chain Reaction/statistics & numerical data , Retina/metabolism
3.
Int J Oncol ; 60(3)2022 Mar.
Article in English | MEDLINE | ID: mdl-35179222

ABSTRACT

Regulator of ribosome synthesis 1 (RRS1) is a key factor in ribosome biosynthesis and other cellular functions. High level of RRS1 in breast cancer cell lines is associated with increased cell proliferation, invasion and migration. RRS1 controls the assembly of the 60s subunit and maturation of 25S rRNA during ribosome biosynthesis. In this study, lentiviral transfection of sh­RNA was used to knock down the level of RRS1, to detect the effect of RRS1 on cell function and to explore the specific mechanism of RRS1 affecting cell invasion and metastasis by COIP and dual­luciferase reporter gene assays. The present study found that RRS1 knockdown reduced the accumulation of ribosome protein L11 (RPL11) in the nucleolus, which then migrated to the nucleoplasm and bound to c­Myc. This inhibited trans­activation of SNAIL by c­Myc and eventually decreased the invasion and metastasis capacity of the human breast cancer cell line BT549. Taken together, RRS1 regulates invasion and metastasis of human breast cancer cells through the RPL11­c­Myc­SNAIL axis. The findings are of great significance for exploring the mechanism of breast cancer invasion and metastasis and the corresponding regulatory factors.


Subject(s)
Down-Regulation/genetics , Neoplasm Metastasis/genetics , RNA-Binding Proteins/drug effects , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cell Proliferation/genetics , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/genetics , Down-Regulation/drug effects , Humans , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/prevention & control , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Snail Family Transcription Factors/drug effects , Snail Family Transcription Factors/genetics , Transcription Factors/drug effects , Transcription Factors/genetics
4.
Anticancer Drugs ; 33(1): e134-e144, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34387591

ABSTRACT

Circular RNAs (circRNAs) are important regulators that drive or inhibit cancer initiation and development. Here, we identified the expression and function of a circRNA, circ_KIAA1199, in colorectal cancer (CRC). The expression levels of circ_KIAA1199, microRNA-34c-5p (miR-34c-5p) and Musashi RNA-binding protein 1 (MSI1) mRNA were detected by quantitative real-time PCR. Cell proliferative capacity was assessed by colony formation assay, EdU assay and MTT assay. Cell apoptosis was determined by flow cytometry assay. Cell migration and cell invasion were investigated by transwell assay. The expression of MSI1 protein and proliferation, migration-related markers was detected by western blot. The relationship between miR-34c-5p and circ_KIAA1199 or MSI1 was verified by dual-luciferase reporter assay. Animal models were constructed to ascertain the role of circ_KIAA1199 in vivo. The expression of circ_KIAA1199 was elevated in CRC. Circ_KIAA1199 downregulation suppressed CRC cell proliferation, survival, migration and invasion. MiR-34c-5p was a target of circ_KIAA1199. The effects of circ_KIAA1199 downregulation were reversed by miR-34c-5p deficiency. In addition, MSI1 was a target of circ_KIAA1199, and the inhibitory effects of miR-34c-5p restoration on CRC cell proliferation, survival, migration and invasion were reversed by MSI1 overexpression. Circ_KIAA1199 positively regulated MSI1 expression by targeting miR-34c-5p. Moreover, circ_KIAA1199 knockdown blocked tumor growth in animal models. Circ_KIAA1199 functioned as an oncogene to drive the malignant development of CRC by activating MSI1 via competitively targeting miR-34c-5p.


Subject(s)
Colorectal Neoplasms/pathology , MicroRNAs/drug effects , Nerve Tissue Proteins/drug effects , RNA, Circular/pharmacology , RNA-Binding Proteins/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Tumor Burden , Xenograft Model Antitumor Assays
5.
JCI Insight ; 6(23)2021 12 08.
Article in English | MEDLINE | ID: mdl-34877932

ABSTRACT

Tristetraprolin (TTP), an important immunosuppressive protein regulating mRNA decay through recognition of the AU-rich elements (AREs) within the 3'-UTRs of mRNAs, participates in the pathogenesis of liver diseases. However, whether TTP regulates mRNA stability through other mechanisms remains poorly understood. Here, we report that TTP was upregulated in acute liver failure (ALF), resulting in decreased mRNA stabilities of CCL2 and CCL5 through promotion of N6-methyladenosine (m6A) mRNA methylation. Overexpression of TTP could markedly ameliorate hepatic injury in vivo. TTP regulated the mRNA stabilization of CCL2 and CCL5. Interestingly, increased m6A methylation in CCL2 and CCL5 mRNAs promoted TTP-mediated RNA destabilization. Moreover, induction of TTP upregulated expression levels of WT1 associated protein, methyltransferase like 14, and YT521-B homology N6-methyladenosine RNA binding protein 2, which encode enzymes regulating m6A methylation, resulting in a global increase of m6A methylation and amelioration of liver injury due to enhanced degradation of CCL2 and CCL5. These findings suggest a potentially novel mechanism by which TTP modulates mRNA stabilities of CCL2 and CCL5 through m6A RNA methylation, which is involved in the pathogenesis of ALF.


Subject(s)
Chemokine CCL2/metabolism , Chemokine CCL5/metabolism , Liver Failure, Acute/drug therapy , Methylation/drug effects , RNA-Binding Proteins/drug effects , Tristetraprolin/therapeutic use , Animals , Humans , Mice , Tristetraprolin/pharmacology
6.
Nat Commun ; 12(1): 6607, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34785661

ABSTRACT

Homeodomain-interacting protein kinases (HIPKs) belong to the CMGC kinase family and are closely related to dual-specificity tyrosine phosphorylation-regulated kinases (DYRKs). HIPKs are regulators of various signaling pathways and involved in the pathology of cancer, chronic fibrosis, diabetes, and multiple neurodegenerative diseases. Here, we report the crystal structure of HIPK3 in its apo form at 2.5 Å resolution. Recombinant HIPKs and DYRK1A are auto-activated and phosphorylate the negative elongation factor SPT5, the transcription factor c-Myc, and the C-terminal domain of RNA polymerase II, suggesting a direct function in transcriptional regulation. Based on a database search, we identified abemaciclib, an FDA-approved Cdk4/Cdk6 inhibitor used for the treatment of metastatic breast cancer, as potent inhibitor of HIPK2, HIPK3, and DYRK1A. We determined the crystal structures of HIPK3 and DYRK1A bound to abemaciclib, showing a similar binding mode to the hinge region of the kinase as observed for Cdk6. Remarkably, DYRK1A is inhibited by abemaciclib to the same extent as Cdk4/Cdk6 in vitro, raising the question of whether targeting of DYRK1A contributes to the transcriptional inhibition and therapeutic activity of abemaciclib.


Subject(s)
Aminopyridines/pharmacology , Benzimidazoles/pharmacology , Gene Expression Regulation/drug effects , Protein Serine-Threonine Kinases/drug effects , Protein-Tyrosine Kinases/drug effects , RNA-Binding Proteins/drug effects , Ribosomal Proteins/drug effects , Breast Neoplasms/drug therapy , Carrier Proteins , Crystallography, X-Ray , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Female , Humans , Intracellular Signaling Peptides and Proteins , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Signal Transduction/drug effects , Dyrk Kinases
7.
Mol Cancer ; 20(1): 78, 2021 05 26.
Article in English | MEDLINE | ID: mdl-34039363

ABSTRACT

BACKGROUND: Distant metastasis is the major cause of death in patients with colorectal cancer (CRC). Previously, we identified KITENIN as a metastasis-enhancing gene and suggested that the oncogenic KITENIN complex is involved in metastatic dissemination of KITENIN-overexpressing CRC cells. Here, we attempted to find substances targeting the KITENIN complex and test their ability to suppress distant metastasis of CRC. METHODS: We screened a small-molecule compound library to find candidate substances suppressing the KITENIN complex in CRC cells. We selected a candidate compound and examined its effects on the KITENIN complex and distant metastasis through in vitro assays, a molecular docking model, and in vivo tumor models. RESULTS: Among several compounds, we identified DKC1125 (Disintegrator of KITENIN Complex #1125) as the best candidate. DKC1125 specifically suppressed KITENIN gain of function. After binding KH-type splicing regulatory protein (KSRP), DKC1125 degraded KITENIN and Dvl2 by recruiting RACK1 and miRNA-124, leading to the disintegration of the functional KITENIN-KSRP-RACK1-Dvl2 complex. A computer docking model suggested that DKC1125 specifically interacted with the binding pocket of the fourth KH-domain of KSRP. KITENIN-overexpressing CRC cells deregulated certain microRNAs and were resistant to 5-fluorouracil, oxaliplatin, and cetuximab. DKC1125 restored sensitivity to these drugs by normalizing expression of the deregulated microRNAs, including miRNA-124. DKC1125 effectively suppressed colorectal liver metastasis in a mouse model. Interestingly, the combination of DKC1125 with 5-fluorouracil suppressed metastasis more effectively than either drug alone. CONCLUSION: DKC1125 targets the KITENIN complex and could therefore be used as a novel therapeutic to suppress liver metastasis in CRC expressing high levels of KITENIN.


Subject(s)
Antineoplastic Agents/pharmacology , Carrier Proteins/drug effects , Colorectal Neoplasms/pathology , Membrane Proteins/drug effects , RNA-Binding Proteins/drug effects , RNA-Binding Proteins/metabolism , Trans-Activators/drug effects , Trans-Activators/metabolism , Animals , Antineoplastic Agents/chemistry , Drug Discovery , Humans , Mice , Molecular Docking Simulation , Neoplasm Metastasis/pathology , RNA-Binding Proteins/antagonists & inhibitors , Trans-Activators/antagonists & inhibitors
8.
Behav Brain Res ; 408: 113295, 2021 06 25.
Article in English | MEDLINE | ID: mdl-33839161

ABSTRACT

AIMS: Considering that serotoninergic agents attenuate symptoms of anxiety and are used to treat depression, we investigated whether subchronic treatment with imipramine, a serotonin/noradrenaline reuptake inhibitor, would prevent the anxiogenic-like behaviour induced by acute and/or chronic ethanol withdrawal. We also investigated whether those changes were related to the disfunctioning of hypothalamic-pituitary-adrenal (HPA) axis and serotonergic neurotransmission. MAIN METHODS: 264 Male Wistar rats were treated with ethanol 6% (vol./vol.) for 21 days. Acute ethanol withdrawal was induced by abrupt discontinuation of treatment and sustained for 48 h. Protracted abstinence was sustained for an additional period of 21 days. Behavioural tests included the Elevated Plus Maze (EPM) or Light/Dark Box (LDB) after acute abstinence, and the Forced Swim Test (FST) after protracted abstinence. Imipramine (15 mg/kg, i.p.) was administered 24, 19 and 1 h before EPM or LDB tests. KEY FINDINGS: Acute abstinence decreased exploration of the open arms of the EPM, without changing exploration of LDB. Additionally, chronic abstinent rats displayed more time immobile in the FST, when compared to control animals. These effects were attenuated by imipramine treatment, without changing basal response. Imipramine prevented protracted abstinence -induced decrease in glucocorticoid receptor (GR) and serotonin transporter (SERT) expression in the dorsal hippocampus. SIGNIFICANCE: Our findings indicate that chronic ethanol withdrawal affects the hippocampal serotonergic system by decreasing serotonin transporter expression. It also disturbs the HPA axis functioning through an imbalance on GR and mineralocorticoid (MR) expression.


Subject(s)
Alcohol Abstinence , Anxiety , Behavior, Animal , Depression , Hippocampus , RNA-Binding Proteins , Receptors, Glucocorticoid , Animals , Anxiety/drug therapy , Anxiety/physiopathology , Behavior, Animal/drug effects , Behavior, Animal/physiology , Depression/drug therapy , Depression/physiopathology , Hippocampus/drug effects , Hippocampus/metabolism , Imipramine , Male , RNA-Binding Proteins/drug effects , RNA-Binding Proteins/metabolism , Rats , Rats, Wistar , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/metabolism
9.
Biomed Pharmacother ; 137: 111419, 2021 May.
Article in English | MEDLINE | ID: mdl-33761622

ABSTRACT

BACKGROUND: Atherosclerosis, inflammatory disease, is a major reason for cardiovascular diseases and stroke. Kaempferol (Kae) has been well-documented to have pharmacological activities in the previous studies. However, the detailed mechanisms by which Kae regulates inflammation, oxidative stress, and apoptosis in Human Umbilical Vein Endothelial Cells (HUVECs) remain unknown. METHODS AND RESULTS: The real-time quantitative polymerase chain reaction (RT-qPCR) was used to measure expression levels of circNOL12, nucleolar protein 12 (NOL12), miR-6873-3p, and Fibroblast growth factor receptor substrate 2 (FRS2) in HUVECs treated with either oxidized low-density lipoprotein (ox-LDL) alone or in combination with Kae. The cells viability was assessed by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl-2H-tetrazol-3-ium bromide (MTT) assay. The inflammation and oxidative stress were assessed by checking inflammatory factors, Reactive Oxygen Species (ROS), Superoxide Dismutase (SOD), and Malondialdehyde (MDA) levels in ox-LDL-induced HUVECs. The apoptotic cells were quantified by flow cytometry assay. The western blot assay was used for measuring protein expression. The interaction relationship between miR-6873-3p and circNOL12 or FRS2 was analyzed by dual-luciferase reporter and RNA pull-down assays. Treatment with Kae could inhibit ox-LDL-induced the upregulation of circNOL12 in HUVECs. Importantly, Kae weakened ox-LDL-induced inflammation, oxidative stress, and apoptosis in HUVECs, which was abolished by overexpression of circNOL12. What's more, miR-6873-3p was a target of circNOL12 in HUVECs, and the upregulation of miR-6873-3p overturned circNOL12 overexpression-induced effects on HUVECs treated with ox-LDL and Kae. FRS2 was negatively regulated by miR-6873-3p in HUVECs. CONCLUSION: Kae alleviated ox-LDL-induced inflammation, oxidative stress, and apoptosis in HUVECs by regulating circNOL12/miR-6873-3p/FRS2 axis.


Subject(s)
Adaptor Proteins, Signal Transducing/drug effects , Endothelial Cells/drug effects , Kaempferols/pharmacology , Membrane Proteins/drug effects , MicroRNAs/drug effects , Nuclear Proteins/drug effects , RNA-Binding Proteins/drug effects , Signal Transduction/drug effects , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Female , Human Umbilical Vein Endothelial Cells , Humans , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
10.
PLoS Pathog ; 17(3): e1009320, 2021 03.
Article in English | MEDLINE | ID: mdl-33662035

ABSTRACT

Humans are frequently exposed to bacterial genotoxins of the gut microbiota, such as colibactin and cytolethal distending toxin (CDT). In the present study, whole genome microarray-based identification of differentially expressed genes was performed in vitro on HT29 intestinal cells while following the ectopic expression of the active CdtB subunit of Helicobacter hepaticus CDT. Microarray data showed a CdtB-dependent upregulation of transcripts involved in positive regulation of autophagy concomitant with the downregulation of transcripts involved in negative regulation of autophagy. CdtB promotes the activation of autophagy in intestinal and hepatic cell lines. Experiments with cells lacking autophagy related genes, ATG5 and ATG7 infected with CDT- and colibactin-producing bacteria revealed that autophagy protects cells against the genotoxin-induced apoptotic cell death. Autophagy induction could also be associated with nucleoplasmic reticulum (NR) formation following DNA damage induced by these bacterial genotoxins. In addition, both genotoxins promote the accumulation of the autophagic receptor P62/SQSTM1 aggregates, which colocalized with foci concentrating the RNA binding protein UNR/CSDE1. Some of these aggregates were deeply invaginated in NR in distended nuclei together or in the vicinity of UNR-rich foci. Interestingly, micronuclei-like structures and some vesicles containing chromatin and γH2AX foci were found surrounded with P62/SQSTM1 and/or the autophagosome marker LC3. This study suggests that autophagy and P62/SQSTM1 regulate the abundance of micronuclei-like structures and are involved in cell survival following the DNA damage induced by CDT and colibactin. Similar effects were observed in response to DNA damaging chemotherapeutic agents, offering new insights into the context of resistance of cancer cells to therapies inducing DNA damage.


Subject(s)
Autophagy/drug effects , Bacterial Toxins/pharmacology , DNA-Binding Proteins/metabolism , RNA-Binding Proteins/drug effects , Sequestosome-1 Protein/metabolism , Autophagy/physiology , Cell Nucleus/metabolism , Endoplasmic Reticulum Stress/physiology , Helicobacter hepaticus/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Mutagens/metabolism , RNA-Binding Proteins/metabolism , Sequestosome-1 Protein/genetics
11.
Biomed Pharmacother ; 137: 111320, 2021 May.
Article in English | MEDLINE | ID: mdl-33578232

ABSTRACT

In China, Baitouweng Tang (BTWT) is a commonly prescribed remedy for the treatment of ulcerative colitis (UC). Herein, the present study aims to assess the anti-colitis activity of BTWT and its underlying mechanisms in UC BALB/c mice. Induction of UC in BALB/c mice was carried out by adding 3.5% DSS in the drinking water of underlined mice. After UC induction, the mice were administrated with BTWT for 7 days. Clinical symptoms were assessed, followed by analyzing the bile acids (BAs) in serum, liver, colon, bile, and feces of UC mice through UPLC-MS/MS. The modified 16S rDNA high-throughput sequencing was carried out to examine the gut microbiota of feces. BTWT significantly improved the clinical symptoms such as and histological injury and colon shortening in UC induced mice. Furthermore, BTWT remarkably ameliorated colonic inflammatory response. After BTWT treatment, the increased concentrations of UDCA, HDCA, αMCA, ßMCA, CA, and GLCA in UC were decreased, and the levels of some BAs, especially CA, αMCA, and ßMCA were normalized. Moreover, the relative species abundance and gut microbiota diversity in the BTWT-exposed groups were found to be considerably elevated than those in the DSS-treated group. BTWT increased the relative abundance of Firmicutes, Proteobacteria, Actinobacteria, Tenericutes, and TM7, which were statistically lower in the fecal microbiota of UC mice. The relative abundance of Bacteroidetes was found to be elevated in the DSS group and normalized after BTWT treatment. BTWT increased the expression of FXR and TGR5 in the liver. BTWT administration improved DSS-induced mice signs by increasing the TGR5 and FXR expression levels. This result was achieved by the regulation of the BAs and gut microbiota.


Subject(s)
Bile Acids and Salts/metabolism , Colitis, Ulcerative/drug therapy , Drugs, Chinese Herbal/therapeutic use , Gastrointestinal Microbiome/drug effects , RNA-Binding Proteins/drug effects , Signal Transduction/drug effects , Animals , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/pathology , Colon/microbiology , Colon/pathology , Dextran Sulfate , Feces/microbiology , Gene Expression Regulation/drug effects , High-Throughput Nucleotide Sequencing , Male , Mice , Mice, Inbred BALB C , RNA, Ribosomal, 16S/genetics , Receptors, G-Protein-Coupled/metabolism
12.
J Cereb Blood Flow Metab ; 41(5): 1080-1090, 2021 05.
Article in English | MEDLINE | ID: mdl-32615885

ABSTRACT

In eukaryotes, the polyamine pathway generates spermidine that activates the hypusination of the translation factor eukaryotic initiation factor 5A (eIF5A). Hypusinated-eIF5A modulates translation, elongation, termination and mitochondrial function. Evidence in model organisms like drosophila suggests that targeting polyamines synthesis might be of interest against ischemia. However, the potential of targeting eIF5A hypusination in stroke, the major therapeutic challenge specific to ischemia, is currently unknown. Using in vitro models of ischemic-related stress, we documented that GC7, a specific inhibitor of a key enzyme in the eIF5A activation pathway, affords neuronal protection. We identified the preservation of mitochondrial function and thereby the prevention of toxic ROS generation as major processes of GC7 protection. To represent a thoughtful opportunity of clinical translation, we explored whether GC7 administration reduces the infarct volume and functional deficits in an in vivo transient focal cerebral ischemia (tFCI) model in mice. A single GC7 pre- or post-treatment significantly reduces the infarct volume post-stroke. Moreover, GC7-post-treatment significantly improves mouse performance in the rotarod and Morris water-maze, highlighting beneficial effects on motor and cognitive post-stroke deficits. Our results identify the targeting of the polyamine-eIF5A-hypusine axis as a new therapeutic opportunity and new paradigm of research in stroke and ischemic diseases.


Subject(s)
Guanine/analogs & derivatives , Lysine/analogs & derivatives , Mitochondria/metabolism , Oxidoreductases Acting on CH-NH Group Donors/antagonists & inhibitors , Peptide Initiation Factors/metabolism , RNA-Binding Proteins/metabolism , Stroke/therapy , Animals , Behavior, Animal/drug effects , Cognition/drug effects , Guanine/administration & dosage , Guanine/pharmacology , Guanine/therapeutic use , Injections, Intraperitoneal , Ischemic Attack, Transient/drug therapy , Ischemic Attack, Transient/prevention & control , Lysine/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Mitochondria/ultrastructure , Models, Animal , Neuroprotection/drug effects , Oxidative Stress/drug effects , Peptide Initiation Factors/drug effects , Polyamines/metabolism , RNA-Binding Proteins/drug effects , Reactive Oxygen Species/toxicity , Stroke/metabolism , Eukaryotic Translation Initiation Factor 5A
13.
Biochimie ; 177: 50-52, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32805303

ABSTRACT

Various interferon (IFN)-inducible transmembrane (IFITM) proteins are known to be expressed in human tissues though only IFITM 1-3 are inducible by IFN. Numerous studies have shown that activation of IFITM3 could suppress infection by influenza and coronaviruses such as the Middle East Respiratory Syndrome Coronavirus (MERS-CoV). In view of the potential application of IFITM proteins' induction to target SARS-CoV-2 infection that causes COVID-19, this article layout insights into the known antiviral mechanisms and therapeutic agents related to IFITM. Blocking viral entry through various mechanisms and the potential application of the FDA approved immunosuppressant agent, mycophenolic acid, as inducer of IFITM3 are among those discussed.


Subject(s)
Betacoronavirus/drug effects , Coronavirus Infections/drug therapy , Interferons/pharmacology , Membrane Proteins/drug effects , Mycophenolic Acid/pharmacology , Pneumonia, Viral/drug therapy , RNA-Binding Proteins/drug effects , Animals , COVID-19 , Coronavirus Infections/immunology , Coronavirus Infections/metabolism , Humans , Immunosuppressive Agents/pharmacology , Membrane Proteins/immunology , Pandemics , Pneumonia, Viral/immunology , Pneumonia, Viral/metabolism , RNA-Binding Proteins/immunology , SARS-CoV-2 , COVID-19 Drug Treatment
14.
Nucleic Acids Res ; 48(15): 8576-8590, 2020 09 04.
Article in English | MEDLINE | ID: mdl-32761127

ABSTRACT

The IGF2 mRNA-binding protein 1 (IGF2BP1) is a non-catalytic post-transcriptional enhancer of tumor growth upregulated and associated with adverse prognosis in solid cancers. However, conserved effector pathway(s) and the feasibility of targeting IGF2BP1 in cancer remained elusive. We reveal that IGF2BP1 is a post-transcriptional enhancer of the E2F-driven hallmark in solid cancers. IGF2BP1 promotes G1/S cell cycle transition by stabilizing mRNAs encoding positive regulators of this checkpoint like E2F1. This IGF2BP1-driven shortening of the G1 cell cycle phase relies on 3'UTR-, miRNA- and m6A-dependent regulation and suggests enhancement of cell cycle progression by m6A-modifications across cancers. In addition to E2F transcription factors, IGF2BP1 also stabilizes E2F-driven transcripts directly indicating post-transcriptional 'super'-enhancer role of the protein in E2F-driven gene expression in cancer. The small molecule BTYNB disrupts this enhancer function by impairing IGF2BP1-RNA association. Consistently, BTYNB interferes with E2F-driven gene expression and tumor growth in experimental mouse tumor models.


Subject(s)
E2F Transcription Factors/genetics , Neoplasms/genetics , RNA-Binding Proteins/genetics , 3' Untranslated Regions/genetics , Animals , Cell Line, Tumor , E2F1 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Neoplasms/pathology , RNA-Binding Proteins/drug effects , Small Molecule Libraries/pharmacology
15.
Pharm Res ; 37(7): 145, 2020 Jul 14.
Article in English | MEDLINE | ID: mdl-32666304

ABSTRACT

PURPOSE: To construct aptamer AS1411-functionalized targeted lipid nanobubbles that could simultaneously target abnormally highly expressed nucleolin (NCL) on tumor tissue and neovasculature. Additionally, the study of their contrast-enhanced ultrasound molecular imaging capabilities in vitro and in vivo to explore new methods and approaches for the early and accurate diagnosis of triple-negative breast cancer (TNBC). METHODS: First, the targeted lipid-nucleic acid molecules were constructed by an amide reaction. Then, the targeted lipid nanobubbles (AS1411-NBs) and nontargeted lipid nanobubbles (NBs) were prepared by membrane hydration, mechanical vibration and centrifugal floatation. The physicochemical characteristics and contrast-enhanced ultrasound imaging capabilities of AS1411-NBs and NBs were compared and analyzed in vitro and in vivo. RESULTS: There were no significant differences between the AS1411-NBs and NBs in their concentration, average particle size or ultrasound imaging capabilities in vitro (P > 0.05). However, AS1411-NBs could simultaneously target NCL in tumor tissue and neovasculature to effectively prolong the duration of contrast-enhanced ultrasound imaging compared to NBs in vivo. The area under the time-intensity curve was significantly different between AS1411-NBs and NBs (P < 0.001), and the drug loading capacity of the AS1411-NBs was also significantly higher than that of the NBs (P < 0.05). CONCLUSIONS: Aptamer AS1411-functionalized targeted lipid nanobubbles could significantly prolong the duration of contrast-enhanced ultrasound imaging to achieve dual-targeted ultrasound molecular imaging of tumor tissue and neovasculature. AS1411-NBs also have higher drug loading and targeted drug delivery capabilities compared with NBs, which can provide new methods and approaches for the early accurate diagnosis and effective treatment of TNBC.


Subject(s)
Breast Neoplasms/diagnostic imaging , Contrast Media/chemistry , Lipids/chemistry , Microbubbles , Phosphoproteins/drug effects , RNA-Binding Proteins/drug effects , Triple Negative Breast Neoplasms/diagnostic imaging , Animals , Cell Line, Tumor , Drug Delivery Systems , Female , Humans , Mice , Mice, Inbred BALB C , Molecular Imaging/methods , Neovascularization, Pathologic/diagnostic imaging , Particle Size , Ultrasonography , Xenograft Model Antitumor Assays , Nucleolin
16.
J Cyst Fibros ; 19(5): 733-741, 2020 09.
Article in English | MEDLINE | ID: mdl-32067958

ABSTRACT

BACKGROUND: Cystic fibrosis (CF) is a recessive disorder caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. We previously described a first-in-class CFTR modulator that functions as an amplifier to selectively increase CFTR expression and function. The amplifier mechanism is distinct from and complementary to corrector and potentiator classes of CFTR modulators. Here we characterize the mechanism by which amplifiers increase CFTR mRNA, protein, and activity. METHODS: Biochemical studies elucidated the action of amplifiers on CFTR mRNA abundance and translation and defined the role of an amplifier-binding protein that was identified using chemical proteomics. RESULTS: Amplifiers stabilize CFTR mRNA through a process that requires only the translated sequence of CFTR and involves translational elongation. Amplifiers enrich ER-associated CFTR mRNA and increase its translational efficiency through increasing the fraction of CFTR mRNA associated with polysomes. Pulldowns identified the poly(rC)-binding protein 1 (PCBP1) as directly binding to amplifier. A PCBP1 consensus element was identified within the CFTR open reading frame that binds PCBP1. This sequence proved necessary for amplifier responsiveness. CONCLUSIONS: Small molecule amplifiers co-translationally increase CFTR mRNA stability. They enhance translation through addressing the inherently inefficient membrane targeting of CFTR mRNA. Amplifiers bind directly to PCBP1, show enhanced affinity in the presence of bound RNA, and require a PCBP1 consensus element within CFTR mRNA to elicit translational effects. These modulators represent a promising new and mechanistically novel class of CFTR therapeutic. They may be useful as a monotherapy or in combination with other CFTR modulators.


Subject(s)
Chloride Channel Agonists/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis/genetics , DNA-Binding Proteins/drug effects , RNA-Binding Proteins/drug effects , Cystic Fibrosis/drug therapy , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , DNA-Binding Proteins/physiology , Gene Expression Regulation/drug effects , Humans , RNA, Messenger/drug effects , RNA-Binding Proteins/physiology
17.
Biomater Sci ; 8(7): 1934-1950, 2020 Mar 31.
Article in English | MEDLINE | ID: mdl-32039412

ABSTRACT

An aptamer-conjugated gold nanostar (dsDDA-AuNS) has been developed for targeting nucleolin present in both tumor cells and tumor vasculature for conducting a drug-resistant cancer therapy. AuNS with its strong absorption in the near-infrared (NIR) region was assembled with a layer of the anti-nucleolin aptamer AS1411. An anticancer drug, namely doxorubicin (DOX), was specifically conjugated on deoxyguanosine residues employing heat and acid labile methylene linkages. In response to NIR irradiation, dsDDA-AuNS allowed on-demand therapeutics. AS1411 played an active role in drug cargo-nucleus interactions, enhancing drug accumulation in the nuclei of drug-resistant breast cancer cells. The intravenous injection of dsDDA-AuNS allowed higher drug accumulation in drug-resistant tumors over naked drugs, leading to greater therapeutic efficacy even at a 54-fold less equivalent drug dose. The in vivo triggered release of DOX from dsDDA-AuNS was achieved by NIR irradiation, resulting in simultaneous photothermal and chemotherapeutic actions, yielding superior tumor growth inhibition than those obtained from either type of monotherapy for overcoming drug resistance in cancers.


Subject(s)
Antineoplastic Agents/administration & dosage , Aptamers, Nucleotide/administration & dosage , Breast Neoplasms/drug therapy , Doxorubicin/administration & dosage , Drug Resistance, Neoplasm/drug effects , Gold/chemistry , Oligodeoxyribonucleotides/administration & dosage , Administration, Intravenous , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/pharmacology , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Doxorubicin/chemistry , Doxorubicin/pharmacology , Female , Humans , MCF-7 Cells , Metal Nanoparticles , Mice , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/pharmacology , Phosphoproteins/drug effects , Phosphoproteins/genetics , RNA-Binding Proteins/drug effects , RNA-Binding Proteins/genetics , Xenograft Model Antitumor Assays , Nucleolin
19.
Psychiatr Genet ; 30(2): 57-59, 2020 04.
Article in English | MEDLINE | ID: mdl-31895084

ABSTRACT

Tardive dystonia is one of the most serious adverse events that can be caused by antipsychotic treatment, but few studies have examined the etiology of tardive dystonia, and no genetic study using a next-generation sequencing technique has been performed to date. We conducted exome sequencing in three subjects with severe tardive dystonia. We analyzed the results focusing on candidate genes of primary dystonia, for example, TOR1A, GCH1, TH, THAP1, and SGCE. There were no single-nucleotide polymorphisms of these dystonia genes that were commonly shared among our subjects. Instead, the results revealed the presence of rare mutations (minor allele frequency <0.01) on the ZNF806 and SART3 genes in all three patients. This is the first study to analyze whole-exonic regions of the genomes of patients with tardive dystonia. These results were only preliminary, but they suggest that subjects presenting with tardive dystonia induced by antipsychotic treatment can have a genetic predisposition to tardive dystonia.


Subject(s)
Antipsychotic Agents/adverse effects , Tardive Dyskinesia/etiology , Tardive Dyskinesia/genetics , Adult , Alleles , Antigens, Neoplasm/drug effects , Antigens, Neoplasm/genetics , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/genetics , Exome/genetics , Gene Frequency/genetics , Genetic Predisposition to Disease/genetics , Humans , Japan , Male , Middle Aged , Mutation/genetics , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide/genetics , RNA-Binding Proteins/drug effects , RNA-Binding Proteins/genetics , Tardive Dyskinesia/metabolism , Exome Sequencing/methods
20.
Eur Rev Med Pharmacol Sci ; 23(18): 7749-7756, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31599400

ABSTRACT

OBJECTIVE: To elucidate the function of miRNA-138-5p in the early diabetic retinopathy (DR) and the potential mechanism. MATERIALS AND METHODS: DR model in rats was first established by streptozotocin (STZ) injection. MiRNA-138-5p expression in rat retinal tissues was determined by quantitative Real Time-Polymerase Chain Reaction (qRT-PCR). Besides, its expression in retinal capillary endothelial cells (EC) and pericytes (RP) was also detected. Cell counting kit-8 (CCK-8) assay was performed to evaluate proliferative potentials of EC and RP cells. The target gene of miRNA-138-5p was predicted by bioinformatics and further confirmed by dual-luciferase reporter gene assay. Rescue experiments were carried out to verify whether the target gene could reverse the regulatory effect of miRNA-138-5p on the proliferation of EC and RP cells. RESULTS: MiRNA-138-5p was lowly expressed in retinal tissues of DR rats, as well as in EC and RP cells. Overexpression of miRNA-138-5p suppressed the proliferative rate of EC and RP cells, and miRNA-138-5p knockdown obtained the opposite trends. NOVA1 was verified to be the target gene of miRNA-138-5p by dual-luciferase reporter gene assay and RIP assay, which was highly expressed in retinal tissues of DR rats, EC, and RP cells. MiRNA-138-5p knockdown markedly upregulated the mRNA and protein levels of NOVA1 in EC and RP cells. Of note, the inhibitory effect of miRNA-138-5p overexpression on proliferative potentials of EC and RP cells was reversed by NOVA1 overexpression. On the contrary, miRNA-138-5p knockdown accelerated their proliferative potentials and was further reversed by NOVA1 knockdown. CONCLUSIONS: MiRNA-138-5p was lowly expressed in retinal tissues of DR rats, as well as in EC and RP cells. MiRNA-138-5p regulates the early DR by promoting cell proliferation via targeting NOVA1.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Diabetic Retinopathy/genetics , MicroRNAs/genetics , Streptozocin/pharmacology , Animals , Antibiotics, Antineoplastic/administration & dosage , Apoptosis , Cell Line, Tumor , Cell Proliferation , China/epidemiology , Computational Biology/methods , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Injections, Intraperitoneal , Neuro-Oncological Ventral Antigen , Pericytes/drug effects , Pericytes/metabolism , Protective Agents , RNA, Messenger/genetics , RNA-Binding Proteins/drug effects , RNA-Binding Proteins/metabolism , Rats , Retina/cytology , Retina/metabolism , Streptozocin/administration & dosage , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...