Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
PLoS Biol ; 21(2): e3001947, 2023 02.
Article in English | MEDLINE | ID: mdl-36757932

ABSTRACT

Congenital hydrocephalus (CH) is a common neurological disorder affecting many newborns. Imbalanced neurogenesis is a major cause of CH. Multiple CH-associated mutations are within the RNA-binding domain of Trim71, a conserved, stem cell-specific RNA-binding protein. How these mutations alter stem cell fate is unclear. Here, we show that the CH-associated mutations R595H and R783H in Trim71 accelerate differentiation and enhance neural lineage commitment in mouse embryonic stem cells (mESCs), and reduce binding to mRNAs targeted by wild-type Trim71, consistent with previous reports. Unexpectedly, however, each mutant binds an ectopic and distinct repertoire of target mRNAs. R595H-Trim71, but not R783H-Trim71 nor wild-type Trim71, binds the mRNA encoding ß-catenin and represses its translation. Increasing ß-catenin by overexpression or treatment with a Wnt agonist specifically restores differentiation of R595H-Trim71 mESCs. These results suggest that Trim71 mutations give rise to unique gain-of-function pathological mechanisms in CH. Further, our studies suggest that disruption of the Wnt/ß-catenin signaling pathway can be used to stratify disease etiology and develop precision medicine approaches for CH.


Subject(s)
Hydrocephalus , beta Catenin , Animals , Mice , beta Catenin/genetics , Gain of Function Mutation , Cell Differentiation/genetics , Mutation/genetics , Hydrocephalus/genetics , Wnt Signaling Pathway/genetics
2.
RNA ; 29(10): 1453-1457, 2023 10.
Article in English | MEDLINE | ID: mdl-37414463

ABSTRACT

RNA-binding proteins (RBPs) are critical regulators of gene expression. An RBP typically binds to multiple mRNAs and modulates their expression. Although loss-of-function experiments on an RBP can infer how it regulates a specific target mRNA, the results are confounded by potential secondary effects due to the attenuation of all other interactions of the target RBP. For example, regarding the interaction between Trim71, an evolutionarily conserved RBP, and Ago2 mRNA, although Trim71 binds to Ago2 mRNA and overexpression of Trim71 represses Ago2 mRNA translation, it is puzzling that AGO2 protein levels are not altered in the Trim71 knockdown/knockout cells. To address this, we adapted the dTAG (degradation tag) system for determining the direct effects of the endogenous Trim71. Specifically, we knocked in the dTAG to the Trim71 locus, enabling inducible rapid Trim71 protein degradation. We observed that following the induction of Trim71 degradation, Ago2 protein levels first increased, confirming the Trim71-mediated repression, and then returned to the original levels after 24 h post-induction, revealing that the secondary effects from the Trim71 knockdown/knockout counteracted its direct effects on Ago2 mRNA. These results highlight a caveat in interpreting the results from loss-of-function studies on RBPs and provide a method to determine the primary effect(s) of RBPs on their target mRNAs.


Subject(s)
Protein Biosynthesis , RNA-Binding Proteins , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
3.
EMBO Rep ; 24(2): e55843, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36573342

ABSTRACT

Congenital hydrocephalus (CH) is a major cause of childhood morbidity. Mono-allelic mutations in Trim71, a conserved stem-cell-specific RNA-binding protein, cause CH; however, the molecular basis for pathogenesis mediated by these mutations remains unknown. Here, using mouse embryonic stem cells as a model, we reveal that the mouse R783H mutation (R796H in human) alters Trim71's mRNA substrate specificity and leads to accelerated stem-cell differentiation and neural lineage commitment. Mutant Trim71, but not wild-type Trim71, binds Lsd1 (Kdm1a) mRNA and represses its translation. Specific inhibition of this repression or a slight increase of Lsd1 in the mutant cells alleviates the defects in stem cell differentiation and neural lineage commitment. These results determine a functionally relevant target of the CH-causing Trim71 mutant that can potentially be a therapeutic target and provide molecular mechanistic insights into the pathogenesis of this disease.


Subject(s)
Hydrocephalus , Tripartite Motif Proteins , Animals , Humans , Mice , Histone Demethylases/genetics , Histone Demethylases/metabolism , Hydrocephalus/genetics , Mouse Embryonic Stem Cells/metabolism , Mutation , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/metabolism
4.
BMC Genomics ; 22(1): 852, 2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34819020

ABSTRACT

BACKGROUND: Idiopathic membranous nephropathy (IMN) is a cause of nephrotic syndrome that is increasing in incidence but has unclear pathogenesis. Urinary peptidomics is a promising technology for elucidating molecular mechanisms underlying diseases. Dysregulation of the proteolytic system is implicated in various diseases. Here, we aimed to conduct urinary peptidomics to identify IMN-related proteases. RESULTS: Peptide fingerprints indicated differences in naturally produced urinary peptide components among 20 healthy individuals, 22 patients with IMN, and 15 patients with other kidney diseases. In total, 1,080 peptide-matched proteins were identified, 279 proteins differentially expressed in the urine of IMN patients were screened, and 32 proteases were predicted; 55 of the matched proteins were also differentially expressed in the kidney tissues of IMN patients, and these were mainly involved in the regulation of proteasome-, lysosome-, and actin cytoskeleton-related signaling pathways. The 32 predicted proteases showed abnormal expression in the glomeruli of IMN patients based on Gene Expression Omnibus databases. Western blot revealed abnormal expression of calpain, matrix metalloproteinase 14, and cathepsin S in kidney tissues of patients with IMN. CONCLUSIONS: This work shown the calpain/matrix metalloproteinase/cathepsin axis might be dysregulated in IMN. Our study is the first to systematically explore the role of proteases in IMN by urinary peptidomics, which are expected to facilitate discovery of better biomarkers for IMN.


Subject(s)
Glomerulonephritis, Membranous , Biomarkers , Glomerulonephritis, Membranous/genetics , Humans , Peptide Hydrolases
5.
Blood ; 132(24): 2564-2574, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30257881

ABSTRACT

The mechanistic target of rapamycin (mTOR) is a central regulator of cellular proliferation and metabolism. Depending on its binding partners, mTOR is at the core of 2 complexes that either promote protein biosynthesis (mTOR complex 1; mTORC1) or provide survival and proliferation signals (mTORC2). Protein biosynthesis downstream of mTORC1 plays an important role in MYC-driven oncogenesis with translation inhibitors garnering increasing therapeutic attention. The germinal center B-cell oncogene UCHL1 encodes a deubiquitinating enzyme that regulates the balance between mTOR complexes by disrupting mTORC1 and promoting mTORC2 assembly. While supporting mTORC2-dependent growth and survival signals may contribute to its role in cancer, the suppression of mTORC1 activity is enigmatic, as its phosphorylation of its substrate 4EBP1 promotes protein biosynthesis. To address this, we used proximity-based proteomics to identify molecular complexes with which UCH-L1 associates in malignant B cells. We identified a novel association of UCH-L1 with the translation initiation complex eIF4F, the target of 4EBP1. UCH-L1 associates with and promotes the assembly of eIF4F and stimulates protein synthesis through a mechanism that requires its catalytic activity. Because of the importance of mTOR in MYC-driven oncogenesis, we used novel mutant Uchl1 transgenic mice and found that catalytic activity is required for its acceleration of lymphoma in the Eµ-myc model. Further, we demonstrate that mice lacking UCH-L1 are resistant to MYC-induced lymphomas. We conclude that UCH-L1 bypasses the need for mTORC1-dependent protein synthesis by directly promoting translation initiation, and that this mechanism may be essential for MYC in B-cell malignancy.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Lymphoma, B-Cell/metabolism , Neoplasm Proteins/metabolism , TOR Serine-Threonine Kinases/metabolism , Ubiquitin Thiolesterase/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/genetics , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Phosphorylation , TOR Serine-Threonine Kinases/genetics , Ubiquitin Thiolesterase/genetics
6.
Methods ; 155: 49-57, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30527764

ABSTRACT

RNA-binding proteins are important players in post-transcriptional regulation, such as modulating mRNA splicing, translation, and degradation under diverse biological settings. Identifying and characterizing the RNA substrates is a critical step in deciphering the function and molecular mechanisms of the target RNA-binding proteins. High-throughput sequencing of the RNA fragments isolated by crosslinking immunoprecipitation (CLIP-seq) is one of the standard techniques to identify the in vivo transcriptome-wide binding sites of the target RNA-binding protein. This method is widely used in functional and mechanistic characterizations of RNA-binding proteins. In this review, we provide several practical considerations on performing and analyzing CLIP-seq experiments. Particularly, we focus on how to perform CLIP-seq experiments on endogenous RNA-binding proteins. In addition, we provide a practical summary on how to choose and use computational pipelines from an increasing number of computational methods and packages that are available for analyzing the sequencing datasets from the CLIP-seq experiments. We hope these practical considerations will facilitate experimental biologists in performing and analyzing CLIP-seq experiment to obtain biologically relevant mechanistic insights.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Immunoprecipitation/methods , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/genetics , RNA/genetics , Transcriptome , Cross-Linking Reagents/chemistry , Datasets as Topic , Humans , RNA/metabolism , RNA-Binding Proteins/metabolism , Sequence Analysis, RNA , Software
7.
Stem Cells ; 35(7): 1760-1772, 2017 07.
Article in English | MEDLINE | ID: mdl-28436570

ABSTRACT

Bre is a conserved cellular protein expressed in various tissues. Its major function includes DNA damage repair and anti-apoptosis. Recent studies indicate that Bre is potentially involved in stem cell differentiation although pathophysiological significance along with the molecular mechanisms is still unclear. Here, we report that Bre protein was substantially expressed in the bone tissue and its expression was highly upregulated during the osteogenic differentiation. To test a hypothesis that Bre plays functional roles in the process of osteogenic differentiation, we examined the expression of Bre in an osteoporosis mouse model. Compared with the normal bone tissue, Bre expression in osteoporotic bone was also significantly reduced. Moreover, knockdown of Bre in the mouse bone marrow mesenchymal cells significantly reduced the expression of osteogenic marker genes, the alkaline phosphatase activity, and the mineralization capacity, while overexpression of Bre greatly promoted the osteogenesis both in vitro and in vivo. Interestingly, we founded that knockdown of Bre led to activation of the p53 signaling pathways exhibited by increased p53, p21, and Mdm2. However, when we inhibited the p53 by siRNA silencing or pifithrin-α, the impaired osteogenesis caused by Bre knockdown was greatly restored. Finally, we found that Bre promoted the Mdm2-mediated p53 ubiquitination and degradation by physically interacting with p53. Taken together, our results revealed a novel function of Bre in osteoblast differentiation through modulating the stability of p53. Stem Cells 2017;35:1760-1772.


Subject(s)
Bone and Bones/metabolism , Mesenchymal Stem Cells/metabolism , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Osteogenesis/genetics , Osteoporosis/genetics , Tumor Suppressor Protein p53/genetics , Animals , Benzothiazoles/pharmacology , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone and Bones/pathology , Cell Differentiation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Disease Models, Animal , Female , Gene Expression Regulation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Osteoporosis/metabolism , Osteoporosis/pathology , Osteoporosis/therapy , Primary Cell Culture , Protein Stability/drug effects , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Tissue Engineering , Tissue Scaffolds , Toluene/analogs & derivatives , Toluene/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism
8.
Virus Genes ; 54(3): 343-350, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29541932

ABSTRACT

Herpes simplex virus 1 (HSV-1) encodes various microRNAs (miRNAs), whose targets are largely unknown. miR-H1 is the first discovered HSV-1 miRNA and is expressed predominantly in productive infection. Here we show that ubiquitin protein ligase E3 component n-recognin 1 (Ubr1) is a cellular target of miR-H1. Ubr1 is a RING-type E3 ubiquitin ligase of the Arg/N-end rule pathway, which causes the degradation of proteins bearing "destabilizing" N-terminal residues, such as neurodegeneration-associated protein fragment ß-amyloid. Using model substrates, we found that miR-H1 significantly repressed the expression and activity of Ubr1. Consequently, miR-H1-mediated Ubr1 silencing resulted in the accumulation of ß-amyloid, which might contribute to the neurodegenerative pathogenesis enhanced by HSV-1. Our results provide novel insights into the mechanism by which HSV-1-encoded miR-H1 functions in neurodegenerative pathogenesis through targeting Ubr1-mediated Arg/N-end rule degradation pathway.


Subject(s)
Amyloid beta-Peptides/metabolism , Herpesvirus 1, Human/physiology , MicroRNAs/physiology , RNA, Viral/physiology , Ubiquitin-Protein Ligases/metabolism , HEK293 Cells , Humans , MicroRNAs/biosynthesis , Proteasome Endopeptidase Complex/metabolism , Proteolysis , RNA, Viral/biosynthesis , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors
9.
J Clin Lab Anal ; 32(6): e22430, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29516551

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is a malignant tumor that severely threatens human health. To date, early detection for HCC patients is particularly significant due to their poor survival rates even after liver resection. METHODS: Therefore, an efficient and sensitive detection method for monitoring liver cancer, multiplex methylation-specific PCR (MSP) coupled with capillary electrophoresis, is developed. RESULTS: Simulations demonstrated that the methylation status of RASSF1A, p16, SFRP1, and ELF could be detected even when DNA equaled or exceeded 12.5 ng simultaneously. Also, its accuracy for methylation detection outweighed polyacrylamide gel electrophoresis (87.5%) and agarose electrophoresis (84.3%), reaching 92.1%. Subsequently, we implemented multiplex MSP with capillary electrophoresis to investigate methylation status of the four tumor suppressor genes in tissue specimens and explore the prognostic value for HCC patients. As the data suggested, multivariate cox regression analysis revealed that the recurrence-free survival of 46 patients was greatly associated with portal vein tumor thrombus (PVTT) and p16 methylation and receiver operating characteristic (ROC) curves demonstrated that the predictive range of portal vein tumor thrombus (PVTT) combined with p16 hypermethylation was more sensitive than that of either PVTT or p16 hypermethylation alone with regard to disease recurrence in patients with HCC, which could be testified as a valuable biomarker in Clinical application. CONCLUSION: Multiplex MSP coupled with capillary electrophoresis has an excellent prospect of clinical application for monitoring early liver cancer and screening valuable biomarkers for prognosis of HCC patients.

10.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 49(1): 1-7, 2018 Jan.
Article in Zh | MEDLINE | ID: mdl-29737080

ABSTRACT

OBJECTIVE: To study the regulation of suppressor of cytokine signaling 3 (SOCS3) expression bythe brother of the regulator of the imprinted site (BORIS) in hepatocellular carcinoma cell. METHODS: The expression of SOCS3 mRNA in HCC cell lines was detected by real-time quantitative PCR (qRT-PCR). The expression of SOCS3 protein in knockdown and overexpression BORIS of HCC cell lines was tested by Western blot. The SOCS3 gene promoter methylation statusin the knockdown and overexpression BORIS of hepatocarcinoma cell lines was detected by using methylation specific PCR (MSP-PCR) method.The potential BORIS binding site of SOCS3promoter region was found by UCSC database analysis.The enrichment of BORIS in SOCS3 promoter region in endogenous high expression BORIS of HCC cells was evaluated by using chromatin immunoprecipitation (ChIP)-qPCR (ChIP-qPCR).The SOCS3 promoter region histone methylation status in the knockdown and overexpression BORIS of HCC was detected by ChIP-qPCR. RESULTS: The expression of SOCS3 mRNA in hepatocellular carcinoma cells was higher and SOCS3 protein expression was down-regulated or up-regulated in the knockdown or overexpression of BORIS mRNA hepatocarcinoma cells,so BORIS has a positive regulatory effect on SOCS3 protein expression in hepatocarcinoma cells. MSP-PCR experiments showed that the SOCS3 promoter in SMMC-7721 and HepG2 cells was unmethylated and knockdown of BORIS did not change the methylation status; the SOCS3 promoter region of Huh7 cells was methylated; after overexpression of BORIS,the SOCS3 promoter region was changed to an unmethylated state; the SOCS3 promoter was unmethylated in HCCLM3,overexpression of BORIS did not alter the methylation status. The ChIP-qPCR assay demonstrated that BORIS specifically binds to the SOCS3 promoter region in HCC cells with high expression of BORIS. Histone methylation assay indicated that knockdown of BORIS reduced BORIS enrichment in the SOCS3 promoter region, with decreasing H3K4 me2 and increasing H3K27 me3 in the region of histone,whereas the overexpress BORIS in HCC cells showed the opposite situation. CONCLUSION: BORIS plays a role of epigenetic regulationon SOCS3 gene promoter methylation and histone methylation,modulating the expression of SOCS3,and then involved in the development of hepatocellular carcinoma.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Methylation , Histones/metabolism , Humans , Promoter Regions, Genetic
11.
Plasmid ; 90: 15-19, 2017 03.
Article in English | MEDLINE | ID: mdl-28189631

ABSTRACT

Lentiviral vectors are now widely considered as one of the most common gene delivery tools for dividing and non-dividing cells. However, insertional mutagenesis has been found in clinical trials with retroviral vectors, which poses a safety risk. The use of non-integrating lentiviral (NIL) vectors, which avoid integration, eliminates the insertional mutagenesis problem. These NIL vectors are unable to mediate stable gene delivery into dividing cells, which makes them of limited use in the clinical practice of gene therapy. In this study, we constructed a NIL vector which harbors the scaffold/matrix attachment region (S/MAR) sequence and a therapeutic gene. NIL retained episomal erythropoietin (EPO) gene expression for 74days in dividing cells both with and without selection. Furthermore, Southern blot analysis showed that the NIL vector was retained extrachromosomally in CHO cells. In conclusion, the NIL vector based on an S/MAR sequence retained the extrachromosomal expression of a therapeutic gene in dividing cells. Our results show that NIL vectors maybe a safe and effective means of gene delivery, which is of potential clinical significance.


Subject(s)
Erythropoietin/genetics , Gene Transfer Techniques , Genetic Vectors/metabolism , Lentivirus/genetics , Plasmids/metabolism , Animals , CHO Cells , Cell Division , Cricetulus , Erythropoietin/metabolism , Gene Expression , Genetic Vectors/chemistry , HEK293 Cells , Humans , Lentivirus/metabolism , Peptide Elongation Factor 1/genetics , Peptide Elongation Factor 1/metabolism , Plasmids/chemistry , Promoter Regions, Genetic , Transgenes
12.
Mol Cancer ; 15(1): 40, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27189061

ABSTRACT

BACKGROUND: Recent studies have suggested that cancer cells contain subpopulations that can initiate tumor growth, self-renew, and maintain tumor cell growth. However, for esophageal cancer cells, the relationship between STAT3, microRNAs and cancer stem cells remains unclear. METHODS: Serum-free culture was used to enrich esophageal cancer stem-like cells (ECSLC). Flow cytometry determined the proportion of ECSLC. qPCR were performed to examine expression level of stemness factors, mesenchymal markers, ATP-binding cassette (ABC) transporters, STAT3, miR-181b, CYLD. Western blot were performed to analyze the expression of STAT3, p-STAT3 and CYLD (cylindromatosis). BALB/c mice xenograft studies were conducted to evaluate the tumorigenicity of enriched ECSLC. Sphere formation assay and colony formation assays were employed to analyze the relationship between STAT3 and miR-181b. Luciferase assays were used to evaluate activity which CYLD is a target of miR-181b. RESULTS: Sphere formation cells (SFCs) with properties of ECSLC were enriched. Enriched SFCs in serum-free suspension culture exhibited cancer stem-like cell properties and increased single-positive CD44 + CD24-, stemness factor, mesenchymal marker expression ABC transporters and tumorigenicity in vivo compared with the parental cells. Additionally, we found that reciprocal activation between STAT3 and miR-181b regulated SFCs proliferation. Moreover, STAT3 directly activated miR-181b transcription in SFCs and miR-181b then potentiated p-STAT3 activity. Luciferase assays indicated that CYLD was a direct and functional target of miR-181b. CONCLUSION: The mutual regulation between STAT3 and miR-181b in SFCs was required for proliferation and apoptosis resistance. STAT3 and miR-181b control each other's expression in a positive feedback loop that regulates SFCs via CYLD pathway. These findings maybe is helpful for targeting ECSLC and providing approach for esophageal cancer treatments.


Subject(s)
Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , 3' Untranslated Regions , Animals , Apoptosis/genetics , Binding Sites , Cell Line, Tumor , Cell Proliferation , Deubiquitinating Enzyme CYLD , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Mice , RNA Interference , Spheroids, Cellular , Tumor Cells, Cultured , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
13.
Mol Cancer ; 14: 56, 2015 Mar 07.
Article in English | MEDLINE | ID: mdl-25890196

ABSTRACT

BACKGROUND: Acute myeloid leukemia (AML) is initiated and maintained by a subset of self-renewing leukemia stem cells (LSCs), which contribute to the progression, recurrence and therapeutic resistance of leukemia. However, the mechanisms underlying the maintenance of LSCs drug resistance have not been fully defined. In this study, we attempted to elucidate the mechanisms of LSCs drug resistance. METHODS: We performed reverse phase protein arrays to analyze the expression of anti-apoptotic proteins in the LSC-enriched leukemia cell line KG-1a. Immuno-blotting, cell viability and clinical AML samples were evaluated to verify the micro-assay results. The characteristics and transcriptional regulation of survivin were analyzed with the relative luciferase reporter assay, mutant constructs, chromatin immuno-precipitation (ChIP), quantitative real-time reverse transcription polymerase chain reaction (RT-qPCR), and western blotting. The levels of Sp1, c-Myc, phospho-extracellular signal-regulated kinase (p-ERK), phospho-mitogen and stress-activated protein kinase (p-MSK) were investigated in paired CD34+ and CD34- AML patient samples. RESULTS: Survivin was highly over-expressed in CD34 + CD38- KG-1a cells and paired CD34+ AML patients compared with their differentiated counterparts. Functionally, survivin contributes to the drug resistance of LSCs, and Sp1 and c-Myc concurrently regulate levels of survivin transcription. Clinically, Sp1 and c-Myc were significantly up-regulated and positively correlated with survivin in CD34+ AML patients. Moreover, Sp1 and c-Myc were further activated by the ERK/MSK mitogen-activated protein kinase (MAPK) signaling pathway, modulating survivin levels. CONCLUSION: Our findings demonstrated that ERK/MSK/Sp1/c-Myc axis functioned as a critical regulator of survivin expression in LSCs, offering a potential new therapeutic strategy for LSCs therapy.


Subject(s)
Drug Resistance, Neoplasm/genetics , Inhibitor of Apoptosis Proteins/genetics , Leukemia, Myeloid, Acute/genetics , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinase Kinases/genetics , Proto-Oncogene Proteins c-myc/genetics , Sp1 Transcription Factor/genetics , Adult , Antigens, CD34/genetics , Cell Line, Tumor , Cell Survival/genetics , Female , HL-60 Cells , Humans , K562 Cells , Male , Neoplastic Stem Cells/metabolism , Signal Transduction/genetics , Survivin , Transcription, Genetic/genetics , U937 Cells , Up-Regulation/genetics
14.
Cell Biol Int ; 39(11): 1234-41, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26032007

ABSTRACT

Appropriate packaging and condensation are critical for eukaryotic chromatin's accommodation and separation during cell division. Human vigilin, a multi-KH-domain nucleic acid-binding protein, is associated with alpha satellites of centromeres. DDP1, a vigilin's homolog, is implicated with chromatin condensation and segregation. The expression of vigilin was previously reported to elevate in highly proliferating tissues and increased in a subset of hepatocellular carcinoma patients. Other studies showed that vigilin interacts with CTCF, contributes to regulation of imprinted genes Igf2/H19, and colocalizes with HP1α on heterochromatic satellite 2 and ß-satellite repeats. These studies indicate that human vigilin might be involved in chromatin remodeling and regular cell growth. To investigate the potential role of human vigilin in cell cycle, the correlations between vigilin and chromosomal condensation and segregation were studied. Depletion of human vigilin by RNA interference in HepG2 cells resulted in chromosome undercondensation and various chromosomal defects during mitotic phase, including chromosome misalignments, lagging chromosomes, and chromosome bridges. Aberrant polyploid nucleus in telophase was also observed. Unlike the abnormal staining pattern of chromosomes, the shape of spindle was normal. Furthermore, the chromatin showed a greater sensitivity to MNase digestion. Collectively, our findings show that human vigilin apparently participates in chromatin condensation and segregation.


Subject(s)
Chromatin/drug effects , Chromosome Segregation/physiology , Chromosomes, Human/physiology , RNA-Binding Proteins/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Cycle/genetics , Centromere/metabolism , Chromatin/metabolism , Chromobox Protein Homolog 5 , Chromosomes, Human/genetics , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mitosis/physiology , RNA Interference , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics
15.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 46(5): 661-6, 2015 Sep.
Article in Zh | MEDLINE | ID: mdl-26619531

ABSTRACT

OBJECTIVE: To investigate the mechanisms of interaction between high-density lipoprotein binding protein (HDLBP)-VIGILIN with other proteins, we cloned VIGILIN cDNA N, KH1-7, KH8-12, KH13-14, and C fragments separately into expression vector, and identify the expressed proteins. METHODS: The recombinant plasmid pDsred2-N1/VIGILIN was used as template to amplify VIGILIN full length, VIGILIN N terminal, KH1- 7, KH8-12, KH13-14, C terminal and recombinated them with pGEX 5X 3. After transformed into E. coli BL21 cells, the recombinants were confirmed by enzyme digestion and sequence analysis. After optimizing the IPTG inducing condition, we induced GST-VIGILIN fusion proteins on the appropriate conditions. RESULT: The recombinant plasmids of pGEX 5X 3/VIGILIN FL, pGEX 5X 3/VIGILIN N terminal, pGEX 5X 3/VIGILIN KH1-7, pGEX 5X 3/VIGILIN KH8-12, pGEX 5X 3/VIGILIN KH13-14, pGEX 5X 3/VIGILIN C terminal were constructed successfully, and induced the GST-VIGILIN fusion proteins. CONCLUSION: pGEX 5X 3/VIGILIN FL, pGEX 5X 3/VIGILIN N terminal, pGEX 5X 3/VIGILIN KH1-7, pGEX SX 3/VIGILIN KH8-12, pGEX 5X 3/ VIGILIN KH13-14, pGEX 5X 3/VIGILIN C terminal recombinant plasmids were constructed successfully, and their corresponding fusion proteins were successfully expressed.


Subject(s)
RNA-Binding Proteins/genetics , Cloning, Molecular , DNA, Complementary/genetics , Escherichia coli , Humans , Lipoproteins, HDL/metabolism , Plasmids , RNA-Binding Proteins/biosynthesis , Recombinant Fusion Proteins/biosynthesis
16.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 45(2): 196-201, 2014 Mar.
Article in Zh | MEDLINE | ID: mdl-24749339

ABSTRACT

OBJECTIVE: To explore the effects of CCCTC-binding factor (CTCF) on human liver cancer stem cells (HepG2) and cell proliferation of HepG2 and Nasopharyngeal carcinoma cell line (CNE1). METHODS: The pEGFP-N1/CTCF, CTCF-shRNA and GFP-shRNA plasmids were constructed and transfected into HepG2 and CNE1 cells, and RT-PCR or Western blot were performed to detect the mRNA or protein levels of CTCF. The subpopulation of CD90+ cancer stem cells in HepG2 cells transfected with CTCF-shRNA plasmid or GFP-shRNA plasmid (as transfection control) were assayed by flow cytometry with the wild type HepG2 cells as control. Proliferation of cells transfected with CTCF-overexpression or CTCF-shRNA plasmid was evaluated by MTT assay. RESULTS: The levels of both mRNA and protein of CTCF were increased in pEGFP-N1/CTCF transfected HepG2 and CNE1 cells compared to that in pEGFP-N1 transfected cells (P < 0.05), and decreased in CTCF-shRNA transfected cells compared to that in cells transfected with GFP-shRNA (P < 0.05). The results of flow cytometry demonstrated that, detection rate of CD90+ cells in cells transfected with CTCF-shRNA plasmid [(1.7330 +/- 0.4177)%] was obviously higher than that of wild-type HepG2 cells [(0.5750 +/- 0.0629)%] and cells transfected with GFP-shRNA plasmid [(0.3500 +/- 0.0866)%] (P < 0.05). The results of MTT analysis showed that, alteration of CTCF had no effect on cancer cell proliferation (P > 0.05). CONCLUSION: CTCF inhibits human liver cancer stem cells but no effect on cell proliferation.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Liver Neoplasms/metabolism , Neoplastic Stem Cells/cytology , Repressor Proteins/metabolism , CCCTC-Binding Factor , Cell Line, Tumor , Flow Cytometry , Hep G2 Cells , Humans , Plasmids , RNA, Messenger , RNA, Small Interfering , Transfection
17.
ACS Appl Mater Interfaces ; 16(13): 15741-15751, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38518114

ABSTRACT

The ex vivo replication of the highly helical and fibril structures of load-bearing soft tissue is a challenging goal for the study of hydrogels. Inspired by nature, we prepared tissue-like physical gels based on curdlan and gelatin by self-assembly. The hybrid gels have a flexible fibril-matrix architecture, and the fibril orientation is highly tunable. The tensile strength of the gels can be tuned from ∼1.1 to ∼16.5 MPa. The coil-helix transition and nanofibril formation process in the self-assembly system was thoroughly investigated. These helical gels exhibit excellent cell compatibility, which supports adhesion and oriented growth of neural cells. Furthermore, the oriented nanofibrils in the gel are found to be associated with an upregulated expression of regeneration-related genes like N-cadherin (Cdh2) and neural growth factor (NGF). Owing to the strength and biomimetic structure, these gels have great potential in tissue engineering applications.


Subject(s)
Gelatin , beta-Glucans , Gelatin/chemistry , Hydrogels/pharmacology , Hydrogels/chemistry , Tissue Engineering
18.
Dev Cell ; 59(8): 979-990.e5, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38458189

ABSTRACT

Argonaute (AGO) proteins are evolutionarily conserved RNA-binding proteins that control gene expression through the small RNAs they interact with. Whether AGOs have regulatory roles independent of RNAs, however, is unknown. Here, we show that AGO1 controls cell fate decisions through facilitating protein folding. We found that in mouse embryonic stem cells (mESCs), while AGO2 facilitates differentiation via the microRNA (miRNA) pathway, AGO1 controls stemness independently of its binding to small RNAs. We determined that AGO1 specifically interacts with HOP, a co-chaperone for the HSP70 and HSP90 chaperones, and enhances the folding of a set of HOP client proteins with intrinsically disordered regions. This AGO1-mediated facilitation of protein folding is important for maintaining stemness in mESCs. Our results demonstrate divergent functions between AGO1 and AGO2 in controlling cellular states and identify an RNA-independent function of AGO1 in controlling gene expression and cell fate decisions.


Subject(s)
Argonaute Proteins , Cell Differentiation , Mouse Embryonic Stem Cells , Protein Folding , Animals , Argonaute Proteins/metabolism , Argonaute Proteins/genetics , Mice , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/cytology , MicroRNAs/genetics , MicroRNAs/metabolism , Eukaryotic Initiation Factors/metabolism , Eukaryotic Initiation Factors/genetics , HSP90 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Cell Lineage
19.
Microorganisms ; 12(5)2024 May 20.
Article in English | MEDLINE | ID: mdl-38792859

ABSTRACT

The vaginal microbiota can be classified into five major community state types (CSTs) based on the bacterial content. However, the link between different CST subtypes and vaginal infection remains unclear. Here, we analyzed 2017 vaginal microbiota samples from women of a reproductive age with vaginal infections that were published in the last decade. We found that L. iners was the most dominant in 34.8% of the vaginal samples, followed by L. crispatus (21.2%). CST I was common in healthy individuals, whereas CST III and IV were associated with dysbiosis and infection. CST III-B, IV-A, IV-B, and IV-C0 were prevalent in patients with bacterial vaginosis (BV). Based on the relative abundance of bacteria at the (sub)genus level, a random forest classifier was developed to predict vaginal infections with an area under the curve of 0.83. We further identified four modules of co-occurring bacterial taxa: L. crispatus, Gardnerella, Prevotella, and Bacteroides. The functional prediction revealed that nucleotide biosynthesis pathways were upregulated in patients with human papilloma virus, and carbohydrate degradation pathways were downregulated in patients with BV. Overall, our study identified the bacterial signatures of healthy and infected vaginal microbiota, providing unique insights into the clinical diagnosis and health status prediction of women of a reproductive age.

20.
Biotechnol Lett ; 35(10): 1707-14, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23690049

ABSTRACT

Chronic exposure to solar radiation is the primary cause of photoaging and benign and malignant skin tumors. A conditioned serum-free medium (SFM) was prepared from umbilical cord mesenchymal stem cells (UC-MSCs) and its anti-photoaging effect, following chronic UV irradiation in vitro and in vivo, was evaluated. UC-MSC SFM had a stimulatory effect on human dermal fibroblast proliferation and reduced UVA-induced cell death. In addition, UC-MSC SFM blocked UVA inhibition of superoxide dismutase activity. Topical application of UC-MSC SFM to mouse skin prior to UV irradiation blocked the inhibition of superoxide dismutase and glutathione peroxidase activities, and prevented the upregulation of malonaldehyde. UC-MSC SFM thus protects against photoaging induced by UVA and UVB radiation and is a promising candidate for skin anti-photoaging treatments.


Subject(s)
Cell Survival/radiation effects , Culture Media, Conditioned/chemistry , Fibroblasts/radiation effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Ultraviolet Rays , Umbilical Cord/cytology , Animals , Cell Proliferation , Cells, Cultured , Culture Media, Serum-Free/chemistry , Fibroblasts/physiology , Glutathione Peroxidase/metabolism , Humans , Malondialdehyde/metabolism , Mice , Skin/enzymology , Skin/metabolism , Skin/radiation effects , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL