Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 121(26): e2320835121, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38900797

ABSTRACT

Upper aerodigestive squamous cell carcinoma (UASCC) is a common and aggressive malignancy with few effective therapeutic options. Here, we investigate amino acid metabolism in this cancer, surprisingly noting that UASCC exhibits the highest methionine level across all human cancers, driven by its transporter LAT1. We show that LAT1 is also expressed at the highest level in UASCC, transcriptionally activated by UASCC-specific promoter and enhancers, which are directly coregulated by SCC master regulators TP63/KLF5/SREBF1. Unexpectedly, unbiased bioinformatic screen identifies EZH2 as the most significant target downstream of the LAT1-methionine pathway, directly linking methionine metabolism to epigenomic reprogramming. Importantly, this cascade is indispensable for the survival and proliferation of UASCC patient-derived tumor organoids. In addition, LAT1 expression is closely associated with cellular sensitivity to inhibition of the LAT1-methionine-EZH2 axis. Notably, this unique LAT1-methionine-EZH2 cascade can be targeted effectively by either pharmacological approaches or dietary intervention in vivo. In summary, this work maps a unique mechanistic cross talk between epigenomic reprogramming with methionine metabolism, establishes its biological significance in the biology of UASCC, and identifies a unique tumor-specific vulnerability which can be exploited both pharmacologically and dietarily.


Subject(s)
Carcinoma, Squamous Cell , Gene Expression Regulation, Neoplastic , Large Neutral Amino Acid-Transporter 1 , Methionine , Methionine/metabolism , Humans , Large Neutral Amino Acid-Transporter 1/metabolism , Large Neutral Amino Acid-Transporter 1/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Cell Line, Tumor , Epigenesis, Genetic , Epigenomics/methods , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Mice , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Animals , Cell Proliferation , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Cellular Reprogramming/genetics
2.
Metab Brain Dis ; 36(1): 53-65, 2021 01.
Article in English | MEDLINE | ID: mdl-33044640

ABSTRACT

Ischemic postconditioning (PostC) conventionally refers to a series of brief blood vessel occlusions and reperfusions, which can induce an endogenous neuroprotective effect and reduce cerebral ischemia/reperfusion (I/R) injury. Depending on the site of adaptive ischemic intervention, PostC can be classified as in situ ischemic postconditioning (ISPostC) and remote ischemic postconditioning (RIPostC). Many studies have shown that ISPostC and RIPostC can reduce cerebral IS injury through protective mechanisms that increase cerebral blood flow after reperfusion, decrease antioxidant stress and anti-neuronal apoptosis, reduce brain edema, and regulate autophagy as well as Akt, MAPK, PKC, and KATP channel cell signaling pathways. However, few studies have compared the intervention methods, protective mechanisms, and cell signaling pathways of ISPostC and RIPostC interventions. Thus, in this article, we compare the history, common intervention methods, neuroprotective mechanisms, and cell signaling pathways of ISPostC and RIPostC.


Subject(s)
Brain/blood supply , Ischemic Postconditioning/methods , Neuroprotection , Reperfusion Injury/prevention & control , Animals , Disease Models, Animal , Signal Transduction/physiology
3.
Am J Drug Alcohol Abuse ; 43(5): 602-608, 2017 09.
Article in English | MEDLINE | ID: mdl-28032807

ABSTRACT

BACKGROUND: Alcohol abuse is a serious health problem worldwide that causes a variety of physical and mental disorders. Research has shown that the brain-derived neurotrophic factor (BDNF) plays an important role in alcohol addiction. The BDNF precursor (proBDNF) exhibits different actions than BDNF through separate receptors and pathways in the central nervous system. However, the effects of proBDNF and BDNF in alcohol addiction are not fully known. OBJECTIVES: The objective was to identify the expression patterns and effects of proBDNF and BDNF after chronic alcohol exposure. METHODS: A total of 40 male adult mice were studied. A mouse psychomotor sensitization (PS) model was established to explore the effects of BDNF and proBDNF treatment following chronic alcohol exposure. Reverse transcription PCR (RT-PCR) was performed to measure mRNA levels for BDNF, TrkB, P75NTR, and sortilin in the prefrontal cortex, hippocampus, and dorsal striatum of Kunming mice after chronic alcohol exposure. RESULTS: In Kunming mice, chronic alcohol exposure up-regulated BDNF and TrkB mRNA levels in the prefrontal cortex, but decreased sortilin and P75 mRNA levels in the dorsal striatum. No changes in mRNA levels were found in other measured brain regions in the alcohol and control groups. CONCLUSION: Chronic alcohol exposure induced the region-specific expression of BDNF and proBDNF and their respective receptors in the brain. These results suggest that BDNF and proBDNF signaling pathways may play major roles in alcohol preference and addiction.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Corpus Striatum/drug effects , Ethanol/administration & dosage , Hippocampus/drug effects , Prefrontal Cortex/drug effects , Receptor, trkB/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Corpus Striatum/metabolism , Hippocampus/metabolism , Male , Mice , Motor Activity/drug effects , Prefrontal Cortex/metabolism , Receptors, Nerve Growth Factor/metabolism , Up-Regulation
4.
Growth Factors ; 34(1-2): 19-32, 2016 02.
Article in English | MEDLINE | ID: mdl-27144323

ABSTRACT

Brain-derived neurotrophic factor (BDNF) plays important roles in neural stem cell (NSC) growth. In this study, we investigated whether BDNF exerts its neurotrophic effects through the Wnt/ß-catenin signaling pathway in human embryonic spinal cord NSCs (hESC-NSCs) in vitro. We found an increase in hESC-NSC growth by BDNF overexpression. Furthermore, expression of Wnt1, Frizzled1 and Dsh was upregulated, whereas GSK-3ß expression was downregulated. In contrast, hESC-NSC growth was decreased by BDNF RNA interference. BDNF, Wnt1 and ß-catenin components were all downregulated, whereas GSK-3ß was upregulated. Next, we treated hESC-NSCs with 6-bromoindirubin-3'-oxime (BIO), a small molecule inhibitor of GSK-3ß. BIO reduced the effects of BDNF upregulation/downregulation on the cell number, soma size and differentiation, and suppressed the effect of BDNF modulation on the Wnt signaling pathway. Our findings suggest that BDNF promotes hESC-NSC growth in vitro through crosstalk with the Wnt/ß-catenin signaling pathway, and that this interaction may be mediated by GSK-3ß.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Cell Proliferation , Embryonic Stem Cells/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Neural Stem Cells/metabolism , Wnt Signaling Pathway , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Embryonic Stem Cells/physiology , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , Humans , Neural Stem Cells/physiology , Up-Regulation , beta Catenin/metabolism
5.
Childs Nerv Syst ; 31(6): 863-72, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25904356

ABSTRACT

PURPOSE: The development of the human embryonic spinal cord is very complicated, and many cell types are involved in the process. However, the morphological characteristics of neuronal and glial cells during the development of the human fetal spinal cord have not been described. We investigated the systemic distributions and expression pattern of the cell type-specific markers Neuron-specific nuclear protein (NeuN) and glial fibrillary acidic protein (GFAP) during the development of the human fetal spinal cord, in order to clarify the detailed developmental changes of neuronal and glial cells in chronological and spatial aspects. METHODS: A total of 35 fetuses, aged 3 weeks to 8 months of gestation (E3W-E8M), were studied. The markers used for immunohistochemical study were NeuN and GFAP. RESULTS: The intracellular makers NeuN and GFAP were widely detected expression in different structures and cells during the development of the human fetal spinal cord, including the following: central canal, neuroepithelial layer, internal limiting membrane, mantle layer, marginal layer, basal plate, alar plate, ependymal layer, gray matter, white matter, neuron, astrocytes, and nerve fibers. However, there was an absence of GFAP in astrocytes during early fetal spinal cord development until E9W, and the appearance of GFAP-positive reactivity was later than that of neurons. CONCLUSIONS: We consider that NeuN and GFAP can be used to identify neuronal and glial cells during the development of the human fetal spinal cord, and their distribution differs both chronologically and spatially. These characteristic expression patterns would give us a clue to better understand the developmental characteristics of the human spinal cord.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Glial Fibrillary Acidic Protein/metabolism , Phosphopyruvate Hydratase/metabolism , Spinal Cord/embryology , Spinal Cord/metabolism , Age Factors , Female , Fetus , Gestational Age , Humans , Male , Spinal Cord/cytology
6.
Carcinogenesis ; 35(2): 292-301, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24064224

ABSTRACT

To further our understanding of the pathobiology of esophageal squamous cell carcinoma (ESCC), we previously performed microRNA profiling that revealed downregulation of miR-200b in ESCC. Using quantitative real-time PCR applied to 88 patient samples, we confirmed that ESCC tumors expressed significantly lower levels of miR-200b compared with the respective adjacent benign tissues (P = 0.003). Importantly, downregulation of miR-200b significantly correlated with shortened survival (P = 0.025), lymph node metastasis (P = 0.002) and advanced clinical stage (P = 0.020) in ESCC patients. Quantitative mass spectrometry identified 57 putative miR-200b targets, including Kindlin-2, previously implicated in the regulation of tumor invasiveness and actin cytoskeleton in other cell types. Enforced expression of miR-200b mimic in ESCC cells led to a decrease of Kindlin-2 expression, whereas transfection of miR-200b inhibitor induced Kindlin-2 expression. Furthermore, transfection of miR-200b mimic or knockdown of Kindlin-2 in ESCC cells decreased cell protrusion and focal adhesion (FA) formation, reduced cell spreading and invasiveness/migration. Enforced expression of Kindlin-2 largely abrogated the inhibitory effects of miR-200b on ESCC cell invasiveness. Mechanistic studies revealed that Rho-family guanosine triphosphatases and FA kinase mediated the biological effects of the miR-200b-Kindlin-2 axis in ESCC cells. To conclude, loss of miR-200b, a frequent biochemical defect in ESCC, correlates with aggressive clinical features. The tumor suppressor effects of miR-200b may be due to its suppression of Kindlin-2, a novel target of miR-200b that modulates actin cytoskeleton, FA formation and the migratory/invasiveness properties of ESCC.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cytoskeleton/metabolism , Esophageal Neoplasms/pathology , Focal Adhesions/physiology , Membrane Proteins/genetics , MicroRNAs/genetics , Neoplasm Proteins/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/mortality , Cell Adhesion/physiology , Cell Movement/physiology , Cell Proliferation , Esophageal Neoplasms/genetics , Esophageal Neoplasms/mortality , Female , Humans , Lymphatic Metastasis , Male , Middle Aged , Mutagenesis, Site-Directed , Mutation/genetics , Neoplasm Invasiveness , Phosphorylation , Prognosis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Survival Rate , Tumor Cells, Cultured
7.
J Pathol ; 231(2): 257-70, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23836524

ABSTRACT

In contrast to the well-recognized loss of adherens junctions in cancer progression, the role of desmosomal components in cancer development has not been well explored. We previously demonstrated that desmocollin-2 (DSC2), a desmosomal cadherin protein, is reduced in oesophageal squamous cell carcinoma (ESCC), and is associated with enhanced tumour metastasis and poor prognosis. Here, we report that restoration of DSC2 in ESCC cells impeded cell migration and invasion both in vitro and in vivo, whereas siRNA-mediated suppression of DSC2 expression increased cell motility. In E-cadherin-expressing ESCC cells, DSC2 restoration strengthened E-cadherin-mediated adherens junctions and promoted the localization of ß-catenin at these junctions, which indirectly inhibited ß-catenin-dependent transcription. These effects of DSC2 were not present in EC109 cells that lacked E-cadherin expression. ESCC patients with tumours that had reduced E-cadherin and negative DSC2 had poorer clinical outcomes than patients with tumours that lacked either E-cadherin or DSC2, implying that the invasive potential of ESCC cells was restricted by both DSC2 and E-cadherin-dependent junctions. Further studies revealed that DSC2 was a downstream target of miR-25. Enhanced miR-25 promoted ESCC cell invasiveness, whereas restoration of DSC2 abolished these effects. Collectively, our work suggests that miR-25-mediated down-regulation of DSC2 promotes ESCC cell aggressiveness through redistributing adherens junctions and activating beta-catenin signalling.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Desmocollins/metabolism , Esophageal Neoplasms/metabolism , MicroRNAs/metabolism , Neoplasm Invasiveness/genetics , Signal Transduction/physiology , beta Catenin/metabolism , Adherens Junctions/genetics , Adherens Junctions/metabolism , Adherens Junctions/pathology , Adult , Aged , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Desmocollins/genetics , Down-Regulation , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Mice , Mice, Nude , MicroRNAs/genetics , Middle Aged , Neoplasm Invasiveness/pathology , Transfection , Transplantation, Heterologous
8.
Chin J Integr Med ; 30(3): 243-250, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37987961

ABSTRACT

OBJECTIVE: To investigate the effects of Danmu Extract Syrup (DMS) on lipopolysaccharide (LPS)-induced acute lung injury (ALI) in mice and explore the mechanism. METHODS: Seventy-two male Balb/C mice were randomly divided into 6 groups according to a random number table (n=12), including control (normal saline), LPS (5 mg/kg), LPS+DMS 2.5 mL/kg, LPS+DMS 5 mL/kg, LPS+DMS 10 mL/kg, and LPS+Dexamethasone (DXM, 5 mg/kg) groups. After pretreatment with DMS and DXM, the ALI mice model was induced by LPS, and the bronchoalveolar lavage fluid (BALF) were collected to determine protein concentration, cell counts and inflammatory cytokines. The lung tissues of mice were stained with hematoxylin-eosin, and the wet/dry weight ratio (W/D) of lung tissue was calculated. The levels of tumor necrosis factor-α (TNF-α), interleukin (IL)-6 and IL-1 ß in BALF of mice were detected by enzyme linked immunosorbent assay. The expression levels of Claudin-5, vascular endothelial (VE)-cadherin, vascular endothelial growth factor (VEGF), phospho-protein kinase B (p-Akt) and Akt were detected by Western blot analysis. RESULTS: DMS pre-treatment significantly ameliorated lung histopathological changes. Compared with the LPS group, the W/D ratio and protein contents in BALF were obviously reduced after DMS pretreatment (P<0.05 or P<0.01). The number of cells in BALF and myeloperoxidase (MPO) activity decreased significantly after DMS pretreatment (P<0.05 or P<0.01). DMS pre-treatment decreased the levels of TNF-α, IL-6 and IL-1 ß (P<0.01). Meanwhile, DMS activated the phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) pathway and reversed the expressions of Claudin-5, VE-cadherin and VEGF (P<0.01). CONCLUSIONS: DMS attenuated LPS-induced ALI in mice through repairing endothelial barrier. It might be a potential therapeutic drug for LPS-induced lung injury.


Subject(s)
Acute Lung Injury , Drugs, Chinese Herbal , Proto-Oncogene Proteins c-akt , Mice , Male , Animals , Proto-Oncogene Proteins c-akt/metabolism , Lipopolysaccharides , Phosphatidylinositol 3-Kinases/metabolism , Interleukin-1beta/metabolism , Vascular Endothelial Growth Factor A/metabolism , Tumor Necrosis Factor-alpha/metabolism , Claudin-5/metabolism , Acute Lung Injury/drug therapy , Acute Lung Injury/chemically induced , Lung/pathology , Interleukin-6/metabolism
9.
Heliyon ; 10(9): e30169, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38699022

ABSTRACT

Nauclea officinalis, as a Chinese medicine in Hainan province, had the effect of treating lower limb ulcers, burn infections. In this paper, we studied the effect of Strictosamide (STR), the main bioactive compound in Nauclea officinals, on wound healing and explored its internal mechanism. Firstly, the wound healing potential of STR was evaluated in a rat model, demonstrating its ability to expedite wound healing, mitigate inflammatory infiltration, and enhance collagen deposition. Additionally, immunofluorescence analysis revealed that STR up-regulated the expression of CD31 and PCNA. Subsequently, target prediction, protein-protein interaction (PPI), gene ontology (GO), and pathway enrichment analyses were used to obtain potential targets, specific biological processes, and molecular mechanisms of STR for the potential treatment of wound healing. Furthermore, molecular docking was conducted to predict the binding affinity between STR and its associated targets. Additionally, in vivo and in vitro experiments confirmed that STR could increase the expression of P-PI3K, P-AKT and P-mTOR by activating the PI3K/AKT signaling pathway. In summary, this study provided a new explanation for the mechanism by which STR promotes wound healing through network pharmacology, suggesting that STR may be a new candidate for treating wound.

10.
J Ovarian Res ; 16(1): 80, 2023 Apr 22.
Article in English | MEDLINE | ID: mdl-37087441

ABSTRACT

BACKGROUND: Standard platinum-based therapy for ovarian cancer is inefficient against ovarian clear cell carcinoma (OCCC). OCCC is a distinct subtype of epithelial ovarian cancer. OCCC constitutes 25% of ovarian cancers in East Asia (Japan, Korea, China, Singapore) and 6-10% in Europe and North America. The cancer is characterized by frequent inactivation of ARID1A and 10% of cases of endometriosis progression to OCCC. The aim of this study was to identify drugs that are either FDA-approved or in clinical trials for the treatment of OCCC. RESULTS: High throughput screening of 166 compounds that are either FDA-approved, in clinical trials or are in pre-clinical studies identified several cytotoxic compounds against OCCC. ARID1A knockdown cells were more sensitive to inhibitors of either mTOR (PP242), dual mTOR/PI3K (GDC0941), ATR (AZD6738) or MDM2 (RG7388) compared to control cells. Also, compounds targeting BH3 domain (AZD4320) and SRC (AZD0530) displayed preferential cytotoxicity against ARID1A mutant cell lines. In addition, WEE1 inhibitor (AZD1775) showed broad cytotoxicity toward OCCC cell lines, irrespective of ARID1A status. CONCLUSIONS: In a selection of 166 compounds we showed that inhibitors of ATR and WEE1 were cytotoxic against a panel of OCCC cell lines. These two drugs are already in other clinical trials, making them ideal candidates for treatment of OCCC.


Subject(s)
Adenocarcinoma, Clear Cell , Ataxia Telangiectasia Mutated Proteins , Ovarian Neoplasms , Protein-Tyrosine Kinases , Female , Humans , Adenocarcinoma, Clear Cell/drug therapy , Adenocarcinoma, Clear Cell/pathology , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Carcinoma, Ovarian Epithelial , Cell Cycle Proteins/metabolism , China , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Protein-Tyrosine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
11.
Cell Death Differ ; 30(2): 527-543, 2023 02.
Article in English | MEDLINE | ID: mdl-36526897

ABSTRACT

Anillin (ANLN) is a mitosis-related protein that promotes contractile ring formation and cytokinesis, but its cell cycle-dependent degradation mechanisms in cancer cells remain unclear. Here, we show that high expression of ANLN promotes cytokinesis and proliferation in esophageal squamous cell carcinoma (ESCC) cells and is associated with poor prognosis in ESCC patients. Furthermore, the findings of the study showed that the deubiquitinating enzyme USP10 interacts with ANLN and positively regulates ANLN protein levels. USP10 removes the K11- and K63-linked ubiquitin chains of ANLN through its deubiquitinase activity and prevents ANLN ubiquitin-mediated degradation. Importantly, USP10 promotes contractile ring assembly at the cytokinetic furrow as well as cytokinesis by stabilizing ANLN. Interestingly, USP10 and the E3 ubiquitin ligase APC/C co-activator Cdh1 formed a functional complex with ANLN in a non-competitive manner to balance ANLN protein levels. In addition, the macrolide compound FW-04-806 (F806), a natural compound with potential for treating ESCC, inhibited the mitosis of ESCC cells by targeting USP10 and promoting ANLN degradation. F806 selectively targeted USP10 and inhibited its catalytic activity but did not affect the binding of Cdh1 to ANLN and alters the balance of the USP10-Cdh1-ANLN complex. Additionally, USP10 expression was positively correlated with ANLN level and poor prognosis of ESCC patients. Overall, targeting the USP10-ANLN axis can effectively inhibit ESCC cell-cycle progression.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Neoplasms/metabolism , Contractile Proteins/metabolism , Ubiquitin/metabolism , Cell Proliferation , Cell Line, Tumor , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
13.
Zhonghua Fu Chan Ke Za Zhi ; 47(2): 96-100, 2012 Feb.
Article in Zh | MEDLINE | ID: mdl-22455739

ABSTRACT

OBJECTIVES: To explore the roles of mean corpuscular volume (MCV), mean corpuscular hemoglobin (MCH) and hemoglobin A(2) (HbA(2)) in the laboratory screening of thalassemia, and to find optimal screening modality for different conditions. METHODS: From September 2008 to May 2011, 1384 subjects underwent thalassemia screening at Department of Obstetrics and Gynecology of Nanfang Hospital. Of them, 1036 cases were diagnosed with thalassemia (408 α-thalassemia, 608 ß-thalassemia, and 20 αß compound thalassemia, thalassemia group) and 348 without thalassemia, non-thalassemia group. All subjects were screened respectively for MCV, MCH and HbA(2). Analyses were performed in all subjects to assess the sensitivity, specificity, positive predictive value, negative predictive value and diagnostic accuracy respectively associated with MCV, MCH and HbA(2) alone, combination of MCV and MCH, and combination of MCV, MCH and HbA(2). RESULTS: (1) In the thalassemia group, the sensitivity of MCV alone was 92.9% (379/408) for α thalassemia, 99.3% (604/608) for ß thalassemia and 100.0% (20/20) for αß compound thalassemia. In the non-thalassemia group, the specificity of MCV alone was 75.0% (261/348). (2) In the thalassemia group, the sensitivity of MCH alone was 92.9% (379/408) in α thalassemia, 99.0% (602/608) in ß thalassemia and 100.0% (20/20) in αß compound thalassemia. In the non-thalassemia group, the specificity of MCH alone was 72.7% (253/348). (3) The sensitivity of Hb A(2) alone was 67.4% (275/408) for α thalassemia, 97.5% (593/608) for ß thalassemia, and 100% (20/20) for αß compound thalassemia while it's specificity was 72.4% (252/348) in the non-thalassemia group. (4) With positive indexes of MCV, MCH and MCV + MCH, when HbA(2) > 3.5% it had a high value in ß-thalassemia screening, but when HbA(2) < 2.5% it had little value in α-thalassemia screening. (5) As a single marker, MCV and MCH had better sensitivity, specificity, positive predictive value, negative predictive value and diagnosis accuracy than HbA(2). MCV + MCH was the best for overall screening, but for ß thalassemia screening, MCV + MCH + HbA(2) was the best. CONCLUSIONS: MCV and MCH are suitable for epidemic screening in a large population, physical examination and premarital check-up. Hb electrophoresis and thalassemia gene diagnosis are recommended for subjects with positive MCV and MCH indexes. Diagnoses of α and ß-thalassemia gene are recommended for pregnant women with positive MCV and MCH indexes.


Subject(s)
Erythrocyte Indices , Hemoglobin A2/analysis , Mass Screening/methods , alpha-Thalassemia/diagnosis , beta-Thalassemia/diagnosis , Adolescent , Adult , Female , Genetic Carrier Screening , Humans , Middle Aged , Phenotype , Predictive Value of Tests , Pregnancy , Prenatal Diagnosis/methods , Retrospective Studies , Sensitivity and Specificity , Young Adult , alpha-Thalassemia/blood , alpha-Thalassemia/genetics , beta-Thalassemia/blood , beta-Thalassemia/genetics
14.
Biosci Rep ; 42(1)2022 01 28.
Article in English | MEDLINE | ID: mdl-34897384

ABSTRACT

Treatment of peripheral nerve regeneration with stem cells (SCs) alone has some limitations. For this reason, we evaluate the efficacy of neurotrophic factors combined with stem cell transplantation in the treatment of sciatic nerve injury (SNI) in rats. PubMed, Cochrane Library, Embase, WanFang, VIP and China National Knowledge Infrastructure databases were retrieved from inception to October 2021, and control experiments on neurotrophic factors combined with stem cells in the treatment of SNI in rats were searched. Nine articles and 551 rats were included in the meta-analysis. The results of meta-analysis confirmed that neurotrophic factor combined with stem cells for the treatment of SNI yielded more effective repair than normal rats with regard to sciatic nerve index, electrophysiological detection index, electron microscope observation index, and recovery rate of muscle wet weight. The conclusion is that neurotrophic factor combined with stem cells is more conducive to peripheral nerve regeneration and functional recovery than stem cells alone. However, due to the limitation of the quality of the included literature, the above conclusions need to be verified by randomized controlled experiments with higher quality and larger samples.


Subject(s)
Nerve Regeneration , Sciatic Nerve , Animals , Nerve Growth Factors , Rats , Stem Cell Transplantation
15.
Brain Res Bull ; 185: 39-48, 2022 07.
Article in English | MEDLINE | ID: mdl-35452749

ABSTRACT

Remote ischemic postconditioning (RIPostC) is a protective procedure for brain damage caused by ischemia/reperfusion (IR), yet the mechanism of this treatment remains to be elucidated. Circular RNAs (circRNAs) are endogenous non-coding RNAs that have recently been recognized to play vital roles in ischemic brain injury. The aim of this study was to explore the role of circRNAs in the protective mechanism of RIPostC and to analyze the circRNA-microRNA (miRNA) regulation network in RIPostC. Nine rats were assigned randomly into three groups (three rats per group): sham, IR, and RIPostC. Their brain tissues were extracted for next-generation RNA sequencing and bioinformatics analysis was performed for two comparisons: sham vs. IR and IR vs. RIPostC. The expression patterns of selected circRNAs and miRNAs were validated by quantitative real-time PCR (qPCR). We detected 82 upregulated and 51 downregulated circRNAs and 137 upregulated and 127 downregulated miRNAs in the IR group compared with the sham group, and 41 upregulated and 100 downregulated circRNAs and 45 upregulated and 64 downregulated miRNAs in the RIPostC group compared with the IR group. The proposed competitive endogenous RNA (ceRNA) network, which included 24 circRNAs, 20 miRNAs, and 145 mRNAs, indicated that the dysregulated circRNAs played important roles in brain IR injury. On the basis of the expression patterns of selected circRNAs, miRNAs, and mRNAs obtained by qPCR, we proposed a circRNA_0002286-miR-124-3p-VLCAD pathway. In PC12 cell, the expression level of miR-124-3p was significantly upregulated when the expression of circRNA_0002286 was repressed and the expression level of VLCAD (very-long chain acyl-CoA dehydrogenase) was significantly downregulated, which suggested that circRNA_0002286 may act as a miRNA sponge for miR-124-3p to regulate the expression of VLCAD. We found that upregulation of circRNA_0002286 attenuated IR injury and was associated with downregulation of miR-124-3p and upregulation of VLCAD. This is the first time that circRNAs have been shown to be closely related to brain IR injury and RIPostC and suggests that targeting the circRNA_0002286-miR-124-3p-VLCAD pathway might attenuate brain IR injury.


Subject(s)
Brain Ischemia , Ischemic Postconditioning , MicroRNAs , Reperfusion Injury , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Animals , Brain/metabolism , Brain Ischemia/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , RNA, Messenger/metabolism , Rats , Reperfusion Injury/genetics
16.
Neural Regen Res ; 17(6): 1299-1309, 2022 Jun.
Article in English | MEDLINE | ID: mdl-34782575

ABSTRACT

During acute reperfusion, the expression profiles of long noncoding RNAs in adult rats with focal cerebral ischemia undergo broad changes. However, whether long noncoding RNAs are involved in neuroprotective effects following focal ischemic stroke in rats remains unclear. In this study, RNA isolation and library preparation was performed for long noncoding RNA sequencing, followed by determining the coding potential of identified long noncoding RNAs and target gene prediction. Differential expression analysis, long noncoding RNA functional enrichment analysis, and co-expression network analysis were performed comparing ischemic rats with and without ischemic postconditioning rats. Rats were subjected to ischemic postconditioning via the brief and repeated occlusion of the middle cerebral artery or femoral artery. Quantitative real-time reverse transcription-polymerase chain reaction was used to detect the expression levels of differentially expressed long noncoding RNAs after ischemic postconditioning in a rat model of ischemic stroke. The results showed that ischemic postconditioning greatly affected the expression profile of long noncoding RNAs and mRNAs in the brains of rats that underwent ischemic stroke. The predicted target genes of some of the identified long noncoding RNAs (cis targets) were related to the cellular response to ischemia and stress, cytokine signal transduction, inflammation, and apoptosis signal transduction pathways. In addition, 15 significantly differentially expressed long noncoding RNAs were identified in the brains of rats subjected to ischemic postconditioning. Nine candidate long noncoding RNAs that may be related to ischemic postconditioning were identified by a long noncoding RNA expression profile and long noncoding RNA-mRNA co-expression network analysis. Expression levels were verified by quantitative real-time reverse transcription-polymerase chain reaction. These results suggested that the identified long noncoding RNAs may be involved in the neuroprotective effects associated with ischemic postconditioning following ischemic stroke. The experimental animal procedures were approved by the Animal Experiment Ethics Committee of Kunming Medical University (approval No. KMMU2018018) in January 2018.

17.
Oncogene ; 41(4): 502-514, 2022 01.
Article in English | MEDLINE | ID: mdl-34782718

ABSTRACT

Bladder cancer (BLCA) is the most common malignant tumor of the urinary system and is characterized by high metastatic rates and poor prognosis. The expression of tight junction protein 1 (TJP1) is associated with bladder cancer invasion; however, the mechanism by which TJP1 affects vasculature remodeling remains unknown. In this study, we found that TJP1 expression correlated with tumor angiogenesis and poor overall survival in clinical samples. Furthermore, TJP1 overexpression promoted tumor angiogenesis in BLCA cells and stimulated recruitment of macrophages to tumors by upregulating CCL2 expression. Mechanistically, TJP1 interacted with TWIST1 and enhanced the transcriptional activity of CCL2. The impairment of tumor angiogenesis caused by knockdown of TJP1 was dramatically rescued by overexpression of TWIST1. Furthermore, TJP1 recruited USP2, which deubiquitinated TWIST1, thereby protecting TWIST1 from proteasome-mediated protein degradation. In conclusion, our results suggest that TJP1 controls angiogenesis in BLCA via TWIST1-dependent regulation of CCL2. We demonstrate that TJP1 functions as a scaffold for the interaction between USP2 and TWIST1 and this may provide potential therapeutic targets in bladder cancer.


Subject(s)
Ubiquitin Thiolesterase/metabolism , Urinary Bladder Neoplasms/genetics , Zonula Occludens-1 Protein/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , Mice, Nude , Transfection , Urinary Bladder Neoplasms/pathology
18.
Ann Transl Med ; 9(22): 1694, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34988203

ABSTRACT

BACKGROUND: The NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome is an important mediator of neuroinflammatory responses that regulates inflammatory injury following cerebral ischemia and may be a potential target. Salidroside (Sal) has good anti-inflammatory effects; however, it remains unclear whether Sal can regulate NLRP3 inflammasome activation through the Toll-like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB) signaling pathway after cerebral ischemia to alleviate inflammatory injury. METHODS: We established an oxygen-glucose deprivation and reoxygenation (OGD/R) model of BV2 cells and a middle cerebral artery occlusion/reperfusion (MCAO/R) rat model. Cell Counting Kit-8 (CCK-8), flow cytometry and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay were used to detect the viability and apoptosis of BV2 cells. Enzyme-linked immunosorbent assay (ELISA) was used to detect the level of inflammatory factors. 2,3,5-triphenyltetrazolium chloride (TTC) staining and modified Neurological Severity Score (mNSS) were used to detect cerebral infarction volume and neurological deficit in rats. Western blot, immunohistochemistry and immunofluorescence staining were used to detect the protein expression levels. RESULTS: Our results showed that Sal increased viability, inhibited lactate dehydrogenase (LDH) release, and reduced apoptosis in OGD/R-induced BV2 cells. Sal reduced the levels of tumor necrosis factor-α (TNF-α), interleukin (IL)-6, and IL-8. Following induction by OGD/R, BV2 cells exhibited NLRP3 inflammasome activation and increased protein levels of NLRP3, apoptosis-associated speck-like protein containing a CARD (ASC), caspase-1, IL-1ß, and IL-18. Protein levels of key TLR4 signaling pathway elements, such as TLR4, myeloid differentiation primary response 88 (MyD88), and phosphorylated nuclear factor kappa B p65 (p-NF-κB p65)/NF-κB p65 were upregulated. Interestingly, it was revealed that Sal could reverse these changes. In addition, TAK242, a specific inhibitor of TLR4, had the same effect as Sal treatment on BV2 cells following induction by OGD/R. In the MCAO/R rat model, Sal was also observed to inhibit NLRP3 inflammasome activation in microglia, reduce cerebral infarction volume, and inhibit apoptosis. CONCLUSIONS: In summary, we found that Sal inhibited NLRP3 inflammasome activation and apoptosis in microglia induced by cerebral ischemia/reperfusion injury by inhibiting the TLR4/NF-κB signaling pathway, thus playing a protective role. Therefore, Sal may be a promising drug for the clinical treatment of ischemic stroke.

19.
Biomed Pharmacother ; 144: 112273, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34700232

ABSTRACT

Neurogenesis in the adult brain is well recognized and plays a critical role in the maintenance of brain function and homeostasis. However, whether neurogenesis also occurs in the adult peripheral nervous system remains unknown. Here, using sensory ganglia (dorsal root ganglia, DRGs) as a model, we show that neurogenesis also occurs in the peripheral nervous system, but in a manner different from that in the central nervous system. Satellite glial cells (SGCs) express the neuronal precursor markers Nestin, POU domain, class 4, transcription factor 1, and p75 pan-neurotrophin receptor. Following sciatic nerve injury, the suppression of endogenous proBDNF by proBDNF antibodies resulted in the transformation of proliferating SGCs into doublecortin-positive cells in the DRGs. Using purified SGCs migrating out from the DRGs, the inhibition of endogenous proBDNF promoted the conversion of SGCs into neuronal phenotypes in vitro. Our findings suggest that SGCs are neuronal precursors, and that proBDNF maintains the SGC phenotype. Furthermore, the suppression of proBDNF signaling is necessary for neuronal phenotype acquisition by SGCs. Thus, we propose that peripheral neurogenesis may occur via the direct conversion of SGCs into neurons, and that this process is negatively regulated by proBDNF.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Ganglia, Spinal/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Neuroglia/metabolism , Peripheral Nerve Injuries/metabolism , Protein Precursors/metabolism , Action Potentials , Animals , Animals, Newborn , Brain-Derived Neurotrophic Factor/genetics , Cell Transdifferentiation , Cells, Cultured , Disease Models, Animal , Doublecortin Protein/metabolism , Female , Ganglia, Spinal/pathology , Ganglia, Spinal/physiopathology , Male , Neural Stem Cells/pathology , Neuroglia/pathology , Peripheral Nerve Injuries/pathology , Peripheral Nerve Injuries/physiopathology , Phenotype , Protein Precursors/genetics , Rats, Sprague-Dawley , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Signal Transduction
20.
Nat Commun ; 12(1): 4362, 2021 07 16.
Article in English | MEDLINE | ID: mdl-34272396

ABSTRACT

Squamous cell carcinomas (SCCs) comprise one of the most common histologic types of human cancer. Transcriptional dysregulation of SCC cells is orchestrated by tumor protein p63 (TP63), a master transcription factor (TF) and a well-researched SCC-specific oncogene. In the present study, both Gene Set Enrichment Analysis (GSEA) of SCC patient samples and in vitro loss-of-function assays establish fatty-acid metabolism as a key pathway downstream of TP63. Further studies identify sterol regulatory element binding transcription factor 1 (SREBF1) as a central mediator linking TP63 with fatty-acid metabolism, which regulates the biosynthesis of fatty-acids, sphingolipids (SL), and glycerophospholipids (GPL), as revealed by liquid chromatography tandem mass spectrometry (LC-MS/MS)-based lipidomics. Moreover, a feedback co-regulatory loop consisting of SREBF1/TP63/Kruppel like factor 5 (KLF5) is identified, which promotes overexpression of all three TFs in SCCs. Downstream of SREBF1, a non-canonical, SCC-specific function is elucidated: SREBF1 cooperates with TP63/KLF5 to regulate hundreds of cis-regulatory elements across the SCC epigenome, which converge on activating cancer-promoting pathways. Indeed, SREBF1 is essential for SCC viability and migration, and its overexpression is associated with poor survival in SCC patients. Taken together, these data shed light on mechanisms of transcriptional dysregulation in cancer, identify specific epigenetic regulators of lipid metabolism, and uncover SREBF1 as a potential therapeutic target and prognostic marker in SCC.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , Head and Neck Neoplasms/metabolism , Kruppel-Like Transcription Factors/metabolism , Lipid Metabolism/genetics , Lung Neoplasms/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Acetylation , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Chromatin Immunoprecipitation Sequencing , Chromatography, Liquid , Epigenomics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Esophageal Neoplasms/genetics , Fatty Acids/biosynthesis , Fatty Acids/metabolism , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Histones/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Lung Neoplasms/genetics , Regulatory Elements, Transcriptional , Signal Transduction/genetics , Sphingolipids/biosynthesis , Sphingolipids/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tandem Mass Spectrometry , Transcription Factors/genetics , Transcriptome/genetics , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL