Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Mol Cell ; 81(16): 3368-3385.e9, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34375583

ABSTRACT

The mechanistic understanding of nascent RNAs in transcriptional control remains limited. Here, by a high sensitivity method methylation-inscribed nascent transcripts sequencing (MINT-seq), we characterized the landscapes of N6-methyladenosine (m6A) on nascent RNAs. We uncover heavy but selective m6A deposition on nascent RNAs produced by transcription regulatory elements, including promoter upstream antisense RNAs and enhancer RNAs (eRNAs), which positively correlates with their length, inclusion of m6A motif, and RNA abundances. m6A-eRNAs mark highly active enhancers, where they recruit nuclear m6A reader YTHDC1 to phase separate into liquid-like condensates, in a manner dependent on its C terminus intrinsically disordered region and arginine residues. The m6A-eRNA/YTHDC1 condensate co-mixes with and facilitates the formation of BRD4 coactivator condensate. Consequently, YTHDC1 depletion diminished BRD4 condensate and its recruitment to enhancers, resulting in inhibited enhancer and gene activation. We propose that chemical modifications of eRNAs together with reader proteins play broad roles in enhancer activation and gene transcriptional control.


Subject(s)
Adenosine/analogs & derivatives , Cell Cycle Proteins/genetics , Nerve Tissue Proteins/genetics , RNA Splicing Factors/genetics , RNA/genetics , Transcription Factors/genetics , Adenosine/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation/genetics , Humans , Methylation , Regulatory Elements, Transcriptional/genetics , Transcriptional Activation/genetics
2.
Mol Cell ; 75(4): 791-806.e8, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31303470

ABSTRACT

YAP/TEAD are nuclear effectors of the Hippo pathway, regulating organ size and tumorigenesis largely through promoter-associated function. However, their function as enhancer regulators remains poorly understood. Through an in vivo proximity-dependent labeling (BioID) technique, we identified YAP1 and TEAD4 protein as co-regulators of ERα on enhancers. The binding of YAP1/TEAD4 to ERα-bound enhancers is augmented upon E2 stimulation and is required for the induction of E2/ERα target genes and E2-induced oncogenic cell growth. Furthermore, their enhancer binding is a prerequisite for enhancer activation marked by eRNA transcription and for the recruitment of the enhancer activation machinery component MED1. The binding of TEAD4 on active ERE-containing enhancers is independent of its DNA-binding behavior, and instead, occurs through protein-tethering trans-binding. Our data reveal a non-canonical function of YAP1 and TEAD4 as ERα cofactors in regulating cancer growth, highlighting the potential of YAP/TEAD as possible actionable drug targets for ERα+ breast cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Estrogens/pharmacology , Muscle Proteins/metabolism , Neoplasm Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Adaptor Proteins, Signal Transducing/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , DNA-Binding Proteins/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Humans , MCF-7 Cells , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/metabolism , Mice , Mice, Nude , Muscle Proteins/genetics , Neoplasm Proteins/genetics , TEA Domain Transcription Factors , Transcription Factors/genetics , YAP-Signaling Proteins
3.
Environ Toxicol ; 39(3): 1140-1162, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37860845

ABSTRACT

Sulforaphane (SFN) has attracted much attention due to its ability on antioxidant, anti-inflammatory, and anti-apoptotic properties, while its functional targets and underlying mechanism of action on brain injury caused by acute carbon monoxide poisoning (ACOP) have not been fully elucidated. Herein, we used a systematic network pharmacology approach to explore the mechanism of SFN in the treatment of brain damage after ACOP. In this study, the results of network pharmacology demonstrated that there were a total of 81 effective target genes of SFN and 36 drug-disease targets, which were strongly in connection with autophagy-animal signaling pathway, drug metabolism, and transcription disorders in cancer. Upon the further biological function and KEGG signaling pathway enrichment analysis, a large number of them were involved in neuronal death, reactive oxygen metabolic processes and immune functions. Moreover, based on the results of bioinformatics prediction associated with multiple potential targets and pathways, the AMP-activated protein kinase (AMPK) signaling pathway was selected to elucidate the molecular mechanism of SFN in the treatment of brain injury caused by ACOP. The following molecular docking analysis also confirmed that SFN can bind to AMPKα well through chemical bonds. In addition, an animal model of ACOP was established by exposure to carbon monoxide in a hyperbaric oxygen chamber to verify the predicted results of network pharmacology. We found that the mitochondrial ultrastructure of neurons in rats with ACOP was seriously damaged, and apoptotic cells increased significantly. The histopathological changes were obviously alleviated, apoptosis of cortical neurons was inhibited, and the number of Nissl bodies was increased in the SFN group as compared with the ACOP group (p < .05). Besides, the administration of SFN could increase the expressions of phosphorylated P-AMPK and MFN2 proteins and decrease the levels of DRP1, Caspase3, and Casapase9 proteins in the brain tissue of ACOP rats. These findings suggest that network pharmacology is a useful tool for traditional Chinese medicine (TCM) research, SFN can effectively inhibit apoptosis, protect cortical neurons from the toxicity of carbon monoxide through activating the AMPK pathway and may become a potential therapeutic strategy for brain injury after ACOP.


Subject(s)
Brain Injuries , Carbon Monoxide Poisoning , Drugs, Chinese Herbal , Isothiocyanates , Sulfoxides , Rats , Animals , Molecular Docking Simulation , Carbon Monoxide , AMP-Activated Protein Kinases , Network Pharmacology , Brain
4.
Am J Emerg Med ; 61: 18-28, 2022 11.
Article in English | MEDLINE | ID: mdl-36029667

ABSTRACT

INTRODUCTION: Carbon monoxide (CO) poisoning can cause serious neurological sequelae. However, there is neither effective treatment strategy nor reliable indicators to determine the prognosis of patients with CO poisoning. The present study aimed to observe the changes of neurological function score, disease severity score, cerebral oxygen utilization (O2UCc), bispectral (BIS) index and neuron-specific enolase (NSE) concentration, and to elucidate the clinical significance of these potential indicators and the neuroprotective effect of mild hypothermia on brain injury in patients with severe acute CO poisoning. MATERIALS AND METHODS: A total of 277 patients with acute severe CO poisoning from 2013 to 2018 were enrolled in our hospital. Patients were divided into three groups according to their body temperature on the day of admission and their willingness to treat: a fever group (n = 78), a normal temperature group (NT group, n = 113), and a mild hypothermia group (MH group, n = 86). All patients were given hyperbaric oxygen therapy, while those in the MH group received additional mild hypothermia treatment. The severity of the disease, the neurobehavioral status, the incidence of delayed encephalopathy after acute carbon monoxide poisoning (DEACMP), and other indicators including BIS, O2UCc, NSE were further evaluated in all patients at given time-points. RESULTS: Mild hypothermia therapy improved the prognosis of patients with CO poisoning, significantly decreased the value of O2UCc and NSE, and up-regulated BIS. The incidence of DEACMP at 6 months was 27% in the fever group, 23% in the NT group, and 8% in the MH group. The values of Glasgow-Pittsburgh coma scale (G-P score), BIS index and NSE were closely related to the occurrence of DEACMP, the cutoff values were 12.41, 52.17 and 35.20 ng/mL, and the sensitivity and specificity were 79.3%, 77.6%, 79.3% and 67.6%, 89.5%, 88.6% in the receiver operating characteristic curve (ROC), respectively. CONCLUSIONS: Early mild hypothermia treatment could significantly reduce the severity of brain injury after CO poisoning, and might be further popularized in clinic. G-P scores, NSE and BIS index can be regarded as the prediction indicators in the occurrence and development of DEACMP. CLINICAL TRIAL REGISTRATION: The study protocol was granted from Qingdao University Research Ethics Committee (Clinical trial registry and ethical approval number: QD81571283).


Subject(s)
Brain Diseases , Brain Injuries , Carbon Monoxide Poisoning , Hypothermia , Neuroprotective Agents , Humans , Carbon Monoxide Poisoning/complications , Carbon Monoxide Poisoning/therapy , Neuroprotection , Carbon Monoxide , Hypothermia/complications , Phosphopyruvate Hydratase , Oxygen , Brain Diseases/etiology , Brain Diseases/therapy
5.
Environ Toxicol ; 37(3): 413-434, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34761859

ABSTRACT

The pathogenesis of brain injury caused by carbon monoxide poisoning (COP) is very complex, and there is no exact and reliable treatment in clinic. In the present study, we screened the therapeutic target and related signal pathway of Salvia Miltiorrhiza for acute COP brain injury, and clarified the pharmacological mechanism of multicomponent, multitarget, and multisignal pathway in Salvia Miltiorrhiza by network pharmacology. To further verify the therapeutic effect of Salvia Miltiorrhiza on acute brain injury based on the results of network analysis, a total of 216 male healthy Sprague Dawley rats were collected in the present study and randomly assigned to a normal control group, a COP group and a Tanshinone IIA sulfonate treatment group (72 rats in each group). The rat model of acute severe COP was established by the secondary inhalation in a hyperbaric oxygen chamber. We found that Salvia Miltiorrhiza had multiple active components, and played a role in treating acute brain injury induced by COP through multiple targets and multiple pathways, among them, MAPK/ERK1/2 signaling pathway was one of the most important. COP can start apoptosis process, activate the MAPK/ERK1/2 signaling pathway, and promote the expression of VEGF-A protein and the formation of brain edema. Tanshinone IIA can effectively inhibit apoptosis, up-regulate the expressions of VEGF-A, P-MEK1/2 and P-ERK1/2 proteins, thereby protect endothelial cells, promote angiogenesis and microcirculation, and finally alleviate brain edema.


Subject(s)
Brain Injuries , Carbon Monoxide Poisoning , Salvia miltiorrhiza , Animals , Brain Injuries/drug therapy , Carbon Monoxide Poisoning/drug therapy , Endothelial Cells , Internet , Male , Rats , Rats, Sprague-Dawley
6.
Environ Toxicol ; 36(9): 1742-1757, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34032369

ABSTRACT

Oxidative stress has been considered as an important cause of neurocyte damage induced by carbon monoxide (CO) poisoning; however, the precise mechanisms are not fully understood. The study aimed to elucidate the molecular mechanism and the neuroprotective effect of targeted regulatory nuclear factor erythroid2-related factor 2 (Nrf2) gene on acute brain injury in CO poisoning rats. An acute CO poisoning rat model was established by CO inhalation in hyperbaric oxygen chamber and followed by the administration of Nrf2 gene-loaded lentivirus. Mitochondrial membrane potential (ΔΨM), the levels of Nrf2, glutamate-cysteine ligase catalytic subunit (GCLC), catalase (CAT) and glutathione peroxidase (GSH-Px), and cell apoptosis were determined in brain tissue in rats. We found that CO poisoning could decrease ΔΨm of cells, slightly increase the expressions of Nrf2 and GCLC at mRNA and protein levels, reduce CAT and GSH-Px, and thus initiate apoptosis process. The Nrf2 gene treatment could obviously enhance the expressions of Nrf2 at mRNA and protein levels, and increase the concentrations of CAT and GSH-Px, maintain the ΔΨm of cells in brain tissue, significantly inhibit cell apoptosis as compared with the CO poisoning group (p < .05). These findings suggest that CO poisoning could induce oxidative stress and impair mitochondrial function of cells in brain tissue. The administration of Nrf2 gene could notably strengthen the antioxidant capacity of cells through regulating the downstream genes of Nrf2/antioxidant responsive element signal pathway, and positively protect cells against brain injury induced by acute severe CO poisoning.


Subject(s)
Brain Injuries , Carbon Monoxide Poisoning , NF-E2-Related Factor 2 , Neuroprotective Agents , Animals , Brain Injuries/chemically induced , Brain Injuries/genetics , Carbon Monoxide Poisoning/genetics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress , Rats
7.
Microb Cell Fact ; 19(1): 62, 2020 Mar 10.
Article in English | MEDLINE | ID: mdl-32156271

ABSTRACT

BACKGROUND: New semi-synthetic aminoglycoside antibiotics generally use chemical modifications to avoid inactivity from pathogens. One of the most used modifications is 3',4'-di-deoxygenation, which imitates the structure of gentamicin. However, the mechanism of di-deoxygenation has not been clearly elucidated. RESULTS: Here, we report that the bifunctional enzyme, GenB4, catalyzes the last step of gentamicin 3',4'-di-deoxygenation via reduction and transamination activities. Following disruption of genB4 in wild-type M. echinospora, its products accumulated in 6'-deamino-6'-oxoverdamicin (1), verdamicin C2a (2), and its epimer, verdamicin C2 (3). Following disruption of genB4 in M. echinospora ΔgenK, its products accumulated in sisomicin (4) and 6'-N-methylsisomicin (5, G-52). Following in vitro catalytic reactions, GenB4 transformed sisomicin (4) to gentamicin C1a (9) and transformed verdamicin C2a (2) and its epimer, verdamicin C2 (3), to gentamicin C2a (11) and gentamicin C2 (12), respectively. CONCLUSION: This finding indicated that in addition to its transamination activity, GenB4 exhibits specific 4',5' double-bond reducing activity and is responsible for the last step of gentamicin 3',4'-di-deoxygenation. Taken together, we propose three new intermediates that may refine and supplement the specific biosynthetic pathway of gentamicin C components and lay the foundation for the complete elucidation of di-deoxygenation mechanisms.


Subject(s)
Anti-Bacterial Agents/chemistry , Bacterial Proteins/metabolism , Gentamicins/chemistry , Micromonospora/enzymology , Oxygen/chemistry , Amination , Bacterial Proteins/genetics , Biosynthetic Pathways , Catalysis , Micromonospora/genetics , Sisomicin/chemistry
8.
RNA Biol ; 17(11): 1590-1602, 2020 11.
Article in English | MEDLINE | ID: mdl-32506995

ABSTRACT

Breast cancer is one of the most prevalent cancers in women worldwide. Through the regulation of many coding and non-coding target genes, oestrogen (E2 or 17ß-oestradiol) and its nuclear receptor ERα play important roles in breast cancer development and progression. Despite the astounding advances in our understanding of oestrogen-regulated coding genes over the past decades, our knowledge on oestrogen-regulated non-coding targets has just begun to expand. Here we leverage epigenomic approaches to systematically analyse oestrogen-regulated long non-coding RNAs (lncRNAs). Similar to the coding targets of ERα, the transcription of oestrogen-regulated lncRNAs correlates with the activation status of ERα enhancers, measured by eRNA production, chromatin accessibility, and the occupancy of the enhancer regulatory components including P300, MED1, and ARID1B. Our 3D chromatin architecture analyses suggest that lncRNAs and their neighbouring E2-resonsive coding genes, exemplified by LINC00160 and RUNX1, might be regulated as a 3D structural unit resulted from enhancer-promoter interactions. Finally, we evaluated the expression levels of LINC00160 and RUNX1 in various types of breast cancer and found that their expression positively correlated with the survival rate in ER+ breast cancer patients, implying that the oestrogen-regulated LINC00160 and its neighbouring RUNX1 might represent potential biomarkers for ER+ breast cancers.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Epigenesis, Genetic , Estrogens/metabolism , Gene Expression Regulation, Neoplastic , RNA, Long Noncoding/genetics , Receptors, Estrogen/metabolism , Apoptosis , Biomarkers, Tumor , Breast Neoplasms/pathology , Cell Line, Tumor , Computational Biology/methods , Enhancer Elements, Genetic , Estrogen Receptor alpha/metabolism , Estrogens/pharmacology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunophenotyping , Response Elements , Signal Transduction , Transcription, Genetic , Transcriptome
9.
Biochim Biophys Acta ; 1849(3): 270-81, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25514493

ABSTRACT

Pokemon, an important proto-oncoprotein, is a transcriptional repressor that belongs to the POK (POZ and Krüppel) family. Smad4, a key component of TGF-ß pathway, plays an essential role in TGF-ß-induced transcriptional responses. In this study, we show that Pokemon can interact directly with Smad4 both in vitro and in vivo. Overexpression of Pokemon decreases TGF-ß-induced transcriptional activities, whereas knockdown of Pokemon increases these activities. Interestingly, Pokemon does not affect activation of Smad2/3, formation of Smads complex, or DNA binding activity of Smad4. TGF-ß1 treatment increases the interaction between Pokemon and Smad4, and also enhances the recruitment of Pokemon to Smad4-DNA complex. In addition, we also find that Pokemon recruits HDAC1 to Smad4 complex but decreases the interaction between Smad4 and p300/CBP. Taken together, all these data suggest that Pokemon is a new partner of Smad4 and plays a negative role in TGF-ß pathway.


Subject(s)
DNA-Binding Proteins/metabolism , Protein Interaction Maps/genetics , Smad4 Protein/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Binding Sites , DNA-Binding Proteins/genetics , Hep G2 Cells , Humans , Signal Transduction/genetics , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Smad4 Protein/genetics , Transcription Factors/genetics , Transcriptional Activation/genetics , Transforming Growth Factor beta/pharmacology
10.
Biochem Biophys Res Commun ; 480(3): 369-374, 2016 Nov 18.
Article in English | MEDLINE | ID: mdl-27769862

ABSTRACT

MicroRNAs (miRNAs) play important roles in the pathogenesis of many types of cancers by negatively regulating gene expression at posttranscriptional level. Here, we identified that miR-543 is up-regulated in gefitinib-resistant non-small cell lung cancer (NSCLC) patients comparing gefitinib-sensitive ones. It promotes NSCLC cell proliferation by negatively regulates its target gene PTEN. In NSCLC cell lines, CCK-8 proliferation assay indicated that the cell proliferation is promoted by miR-543 mimics. Transwell assay showed that miR-543 mimics promotes the invasion and migration of NSCLC cells. Luciferase assays confirmed that miR-543 directly binds to the 3'untranslated region of PTEN, and western blotting showed that miR-543 suppresses the expression of PTEN at the protein level. This study indicates that miR-543 promotes proliferation and invasion of NSCLC cell lines by PTEN. The miR-543 may represent a potential therapeutic target for gefitinib-resistant NSCLC intervention.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , MicroRNAs/genetics , PTEN Phosphohydrolase/genetics , A549 Cells , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Drug Resistance, Neoplasm/genetics , Enzyme Activation/drug effects , Enzyme Activation/genetics , Female , Gefitinib , Humans , Lung Neoplasms/drug therapy , Male , Middle Aged , Neoplasm Invasiveness , Quinazolines/therapeutic use
11.
Transgenic Res ; 25(5): 693-709, 2016 10.
Article in English | MEDLINE | ID: mdl-27229510

ABSTRACT

Follicle-stimulating hormone (FSH) is a critical hormone regulating reproduction in mammals. Transgenic mice show that overexpression of FSH can improve female fecundity. Using a bacterial artificial chromosome (BAC) system and somatic cell nuclear transfer, we herein generated 67 Large White transgenic (TG) boars harboring FSHα/ß genes from Chinese Erhualian pigs, the most prolific breed in the world. We selected two F0 TG boars for further breeding and conducted molecular characterization and biosafety assessment for F1 boars. We showed that 8-9 copies of exogenous FSHα and 5-6 copies of exogenous FSHß were integrated into the genome of transgenic pigs. The inheritance of exogenous genes conforms to the Mendel's law of segregation. TG boars had higher levels of serum FSH, FSHα mRNA in multiple tissues, FSHß protein in pituitary and more germ cells per seminiferous tubule compared with their wild-type half sibs without any reproductive defects. Analysis of growth curve, hematological and biochemical parameters and histopathology illustrated that TG boars grew healthily and normally. By applying 16S rRNA gene sequencing, we demonstrated that exogenous genes had no impact on the bacterial community structures of pig guts. Moreover, foreign gene drift did not occur as verified by horizontal gene transfer. Our findings indicate that overexpression of FSH could improve spermatogenesis ability of boars. This work provides insight into the effect of FSHα/ß genes on male reproductive performance on pigs by a BAC-mediated transgenic approach.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , Follicle Stimulating Hormone/genetics , Gastrointestinal Microbiome/genetics , Sus scrofa/genetics , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/growth & development , Breeding , Female , Genome , Male , Nuclear Transfer Techniques , RNA, Ribosomal, 16S/genetics , Reproduction/genetics , Spermatogenesis/genetics , Sus scrofa/growth & development
13.
Elife ; 122023 03 07.
Article in English | MEDLINE | ID: mdl-36880874

ABSTRACT

Cerebral ischaemia‒reperfusion injury (IRI), during which neurons undergo oxygen-glucose deprivation/reoxygenation (OGD/R), is a notable pathological process in many neurological diseases. N1-methyladenosine (m1A) is an RNA modification that can affect gene expression and RNA stability. The m1A landscape and potential functions of m1A modification in neurons remain poorly understood. We explored RNA (mRNA, lncRNA, and circRNA) m1A modification in normal and OGD/R-treated mouse neurons and the effect of m1A on diverse RNAs. We investigated the m1A landscape in primary neurons, identified m1A-modified RNAs, and found that OGD/R increased the number of m1A RNAs. m1A modification might also affect the regulatory mechanisms of noncoding RNAs, e.g., lncRNA-RNA binding proteins (RBPs) interactions and circRNA translation. We showed that m1A modification mediates the circRNA/lncRNA‒miRNA-mRNA competing endogenous RNA (ceRNA) mechanism and that 3' untranslated region (3'UTR) modification of mRNAs can hinder miRNA-mRNA binding. Three modification patterns were identified, and genes with different patterns had intrinsic mechanisms with potential m1A-regulatory specificity. Systematic analysis of the m1A landscape in normal and OGD/R neurons lays a critical foundation for understanding RNA modification and provides new perspectives and a theoretical basis for treating and developing drugs for OGD/R pathology-related diseases.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Animals , Mice , RNA, Circular/genetics , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , 3' Untranslated Regions , Glucose , Neurons , Oxygen
14.
Transgenic Res ; 20(2): 417-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20549346

ABSTRACT

In the swine industry pathogenic infections have a significant negative impact on neonatal survival. Piglets fed with human lysozyme, a natural antibiotic, might be more resistant to gastrointestinal infections. Here we describe the generation of transgenic swine expressing recombinant human lysozyme by somatic cell nuclear transfer. Three cloned female pigs were born, one of which expressed rhLZ at 0.32 ± 0.01 µg/ml in milk, 50-fold higher than that of the pig native lysozyme. Both the transgenic gilts and their progeny appear healthy. Introducing human lysozyme into pigs' milk has a potential to benefit the piglets by enhancing immune function and defending against pathogenic bacteria, thereby increasing the new born survival rate. This advance could be of great value to commercial swine producers.


Subject(s)
Animals, Genetically Modified , Milk/enzymology , Muramidase/biosynthesis , Recombinant Proteins/biosynthesis , Swine/genetics , Animal Husbandry/methods , Animals , Cloning, Organism , Female , Gene Transfer Techniques , Humans , Mammary Glands, Animal/metabolism , Muramidase/genetics , Nuclear Transfer Techniques , Recombinant Proteins/genetics , Swine/growth & development , Swine/metabolism , Transgenes
15.
J Ophthalmol ; 2021: 6287083, 2021.
Article in English | MEDLINE | ID: mdl-33532092

ABSTRACT

[This corrects the article DOI: 10.1155/2020/9537360.].

16.
Complement Ther Med ; 52: 102448, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32951711

ABSTRACT

PURPOSE: We aim to analyze the feasibility of external application of Xiao-Shuan-Santo prevent peripherally inserted central catheter (PICC) -related thrombosis. METHODS: A total of 218 patients with PICC catheterization were randomly divided into a control group (n = 103) and a treatment group (n = 115). Patients in the treatment group received additional external application of Xiao-Shuan-San. The changes of coagulation index, the incidence of PICC-related thrombosis and other complications, and the maximum blood flow rate (Vmax) of axillary vein were observed at 1 day before catheterization and 30 days after PICC. RESULTS: At 30 days after PICC, the incidence of PICC-related thrombosis and other adverse events in the treatment group were obviously lower than that in the control group (P < 0.05), and the decreased Vmax value of axillary vein in the control group (11.75±1.91 cm/s) was more visible than that in the treatment group (14.63±3.03 cm/s), accompanied by a statistical significance (P < 0.05). CONCLUSIONS: External application of Xiao-Shuan-San could reduce the incidence of PICC-related thrombosis and other complications.


Subject(s)
Catheterization, Peripheral/adverse effects , Drugs, Chinese Herbal/therapeutic use , Thrombosis/prevention & control , Administration, Topical , Feasibility Studies , Female , Humans , Male , Middle Aged
17.
J Ophthalmol ; 2020: 9537360, 2020.
Article in English | MEDLINE | ID: mdl-33029389

ABSTRACT

PURPOSE: The aim of the present study was to analyze the clinical characteristics of visual dysfunction in patients with carbon monoxide (CO) poisoning. METHODS: A total of 436 patients with CO poisoning were enrolled in our hospital from October 2012 to December 2018, including 193 patients with moderate poisoning (MP group), 165 with severe poisoning (SP group), and 78 with delayed encephalopathy (DE group). The clinical characteristics of visual dysfunction in patients with CO poisoning were analyzed through the collection of medical history, regular physical examination, brain magnetic resonance imaging (MRI), ophthalmological examination, the National Eye Institute Visual Function Questionnaire (NEI-VFQ), and its influencing factors. RESULTS: Some patients in the three groups had visual dysfunction. The main ocular symptoms were local pain, eye movement disorder, and visual field defect. The key pathological factors were keratopathy, retinal nerve cell damage, optic nerve damage, retinal vascular disease, macular disease, and occipital visual center damage. The clinical symptoms of visual dysfunction after CO poisoning lasted for a long time (>12 months) and were not completely consistent with the positive results of the ophthalmological examination. A few sequelae of ophthalmology were still left after the help of medicine. CONCLUSION: The incidence of visual dysfunction in patients with CO poisoning was high, the clinical symptoms were rich and diverse, the duration of disease was long, and the prognosis was poor. Thus, the relevant ophthalmological examination and intervention treatment should be perfected as soon as possible.

18.
Cell Rep ; 31(12): 107803, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32579929

ABSTRACT

The function of enhancer RNAs (eRNAs) in transcriptional regulation remains obscure. By analyzing the genome-wide nascent transcript profiles in breast cancer cells, we identify a special group of eRNAs that are essential for estrogen-induced transcriptional repression. Using eRNAs of TM4SF1 and EFEMP1 as the paradigms, we find that these RNA molecules not only stabilize promoter-enhancer interactions but also recruit liganded estrogen receptor α (ERα) to particular enhancer regions, facilitate the formation of a functional transcriptional complex, and cause gene silencing. Interestingly, ERα is shown to directly bind with eRNAs by its DNA-binding domain. These eRNAs help with the formation of a specific ERα-centered transcriptional complex and promote the association of the histone demethylase KDM2A, which dismisses RNA polymerase II from designated enhancers and suppresses the transcription of target genes. Our work demonstrates a complete mechanism underlying the action of eRNAs in modulating and refining the locus-specific transcriptional program.


Subject(s)
Enhancer Elements, Genetic , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , RNA/metabolism , Cell Line , Down-Regulation/genetics , Estrogen Receptor alpha/chemistry , F-Box Proteins/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Models, Biological , Open Reading Frames/genetics , Protein Binding , Protein Domains , RNA Polymerase II/metabolism , Transcription, Genetic
19.
Nat Cell Biol ; 22(6): 701-715, 2020 06.
Article in English | MEDLINE | ID: mdl-32424275

ABSTRACT

Acquired therapy resistance is a major problem for anticancer treatment, yet the underlying molecular mechanisms remain unclear. Using an established breast cancer cellular model, we show that endocrine resistance is associated with enhanced phenotypic plasticity, indicated by a general downregulation of luminal/epithelial differentiation markers and upregulation of basal/mesenchymal invasive markers. Consistently, similar gene expression changes are found in clinical breast tumours and patient-derived xenograft samples that are resistant to endocrine therapies. Mechanistically, the differential interactions between oestrogen receptor α and other oncogenic transcription factors, exemplified by GATA3 and AP1, drive global enhancer gain/loss reprogramming, profoundly altering breast cancer transcriptional programs. Our functional studies in multiple culture and xenograft models reveal a coordinated role of GATA3 and AP1 in re-organizing enhancer landscapes and regulating cancer phenotypes. Collectively, our study suggests that differential high-order assemblies of transcription factors on enhancers trigger genome-wide enhancer reprogramming, resulting in transcriptional transitions that promote tumour phenotypic plasticity and therapy resistance.


Subject(s)
Adaptation, Physiological , Breast Neoplasms/drug therapy , Cellular Reprogramming , Drug Resistance, Neoplasm , GATA3 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Transcription Factor AP-1/metabolism , Animals , Antineoplastic Agents, Hormonal/pharmacology , Apoptosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , GATA3 Transcription Factor/genetics , Humans , Mice , Mice, Nude , Tamoxifen/pharmacology , Transcription Factor AP-1/genetics , Transcriptional Activation , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL