Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Proc Natl Acad Sci U S A ; 120(21): e2215155120, 2023 05 23.
Article in English | MEDLINE | ID: mdl-37192170

ABSTRACT

Chemistry-alone approach has recently been applied for incepting pluripotency in somatic cells, representing a breakthrough in biology. However, chemical reprogramming is hampered by low efficiency, and the underlying molecular mechanisms remain unclear. Particularly, chemical compounds do not have specific DNA-recognition domains or transcription regulatory domains, and then how do small molecules work as a driving force for reinstating pluripotency in somatic cells? Furthermore, how to efficiently clear materials and structures of an old cell to prepare the rebuilding of a new one? Here, we show that small molecule CD3254 activates endogenous existing transcription factor RXRα to significantly promote mouse chemical reprogramming. Mechanistically, CD3254-RXRα axis can directly activate all the 11 RNA exosome component genes (Exosc1-10 and Dis3) at transcriptional level. Unexpectedly, rather than degrading mRNAs as its substrates, RNA exosome mainly modulates the degradation of transposable element (TE)-associated RNAs, particularly MMVL30, which is identified as a new barrier for cell-fate determination. In turn, MMVL30-mediated inflammation (IFN-γ and TNF-α pathways) is reduced, contributing to the promotion of successful reprogramming. Collectively, our study provides conceptual advances for translating environmental cues into pluripotency inception, particularly, identifies that CD3254-RXRα-RNA exosome axis can promote chemical reprogramming, and suggests modulation of TE-mediated inflammation via CD3254-inducible RNA exosome as important opportunities for controlling cell fates and regenerative medicine.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells , Mice , Animals , Cellular Reprogramming/genetics , Transcription Factors/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , Coumaric Acids/metabolism , Induced Pluripotent Stem Cells/metabolism
2.
EMBO J ; 40(11): e106771, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33909912

ABSTRACT

Chemical compounds have recently been introduced as alternative and non-integrating inducers of pluripotent stem cell fate. However, chemical reprogramming is hampered by low efficiency and the molecular mechanisms remain poorly characterized. Here, we show that inhibition of spleen tyrosine kinase (Syk) by R406 significantly promotes mouse chemical reprogramming. Mechanistically, R406 alleviates Syk / calcineurin (Cn) / nuclear factor of activated T cells (NFAT) signaling-mediated suppression of glycine, serine, and threonine metabolic genes and dependent metabolites. Syk inhibition upregulates glycine level and downstream transsulfuration cysteine biosynthesis, promoting cysteine metabolism and cellular hydrogen sulfide (H2 S) production. This metabolic rewiring decreased oxidative phosphorylation and ROS levels, enhancing chemical reprogramming. In sum, our study identifies Syk-Cn-NFAT signaling axis as a new barrier of chemical reprogramming and suggests metabolic rewiring and redox homeostasis as important opportunities for controlling cell fates.


Subject(s)
Fibroblasts/metabolism , Hydrogen Sulfide/metabolism , Syk Kinase/antagonists & inhibitors , Animals , Calcineurin/metabolism , Cells, Cultured , Cysteine/metabolism , Fibroblasts/drug effects , Glycine/metabolism , Mice , NFATC Transcription Factors/metabolism , Oxazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction
3.
FASEB J ; 38(2): e23406, 2024 01 31.
Article in English | MEDLINE | ID: mdl-38193601

ABSTRACT

Cancer-associated fibroblast (CAF) has emerged as a key contributor to the remodeling of tumor microenvironment through the expression and secretion of extracellular matrix (ECM) proteins, thereby promoting carcinogenesis. However, the precise contribution of ECM proteins from CAFs to gastric carcinogenesis remains poorly understood. In this study, we find that matrilin-3 (MATN3), an upregulated ECM protein associated with poorer prognosis in gastric cancer patients, originates from CAFs in gastric cancer tissues. Ectopic expression of MATN3 in CAFs significantly promotes the invasion of gastric cancer cells, which can be attenuated by neutralizing MATN3 with its antibody. Notably, a portion of MATN3 protein is found to form puncta in gastric cancer tissues ECM. MATN3 undergoes phase separation, which is mediated by its low complexity (LC) and coiled-coil (CC) domains. Moreover, overexpression of MATN3 deleted with either LC or CC in CAFs is unable to promote the invasion of gastric cancer cells, suggesting that LC or CC domain is required for the effect of CAF-secreted MATN3 in gastric cancer cell invasion. Additionally, orthotopic co-injection of gastric cancer cells and CAFs expressing MATN3, but not its ΔLC and ΔCC mutants, leads to enhanced gastric cancer cell invasion in mouse models. Collectively, our works suggest that MATN3 is secreted by CAFs and undergoes phase separation, which promotes gastric cancer invasion.


Subject(s)
Cancer-Associated Fibroblasts , Matrilin Proteins , Stomach Neoplasms , Animals , Humans , Mice , Carcinogenesis , Matrilin Proteins/genetics , Neoplasm Invasiveness , Phase Separation , Stomach Neoplasms/genetics , Tumor Microenvironment
4.
J Gene Med ; 26(1): e3643, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38044747

ABSTRACT

BACKGROUND: Programmed cell death (PCD) has been widely investigated in various human diseases. The present study aimed to identify a novel PCD-related genetic signature in cervical squamous cell carcinoma (CESC) to provide clues for survival, immunotherapy and drug sensitization prediction. METHODS: Single-sample gene set enrichment analysis (ssGSEA) was used to quantify the PCD score and assess the distribution of PCD in clinicopathological characteristics in The Cancer Genome Atlas (TCGA)-CESC samples. Then, the ConsensusClusterPlus method was used to identify molecular subtypes in the TCGA-CESC database. Genomic mutation analysis, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes functional enrichment, as well as tumor microenvironment (TME) infiltration analysis, were performed for each molecular subtype group. Finally, a prognostic model by Uni-Cox and least absolute shrinkage and selection operator-Cox analysis was established based on differentially expressed genes from molecular subtypes. ESTIMATE (i.e. Estimation of STromal and Immune cells in MAlignantTumours using Expression data) and ssGSEA were performed to assess the correlation between the model and TME. Drug sensitization prediction was carried out with the oncoPredict package. RESULTS: Preliminary analysis indicated that PCD had a potential association clinical characteristics of the TCGA-CESC cohort, and PCD-related genes mutated in 289 (70.59%) CESC patients. Next, four groups of CESC molecular typing were clustered based on 63 significantly prognostic PCD-related genes. Among four subtypes, C1 group displayed the worst prognosis combined with over expressed PCD genes and enriched cell cycle-related pathways. C4 group exhibited the best prognosis accompanied with high degree of immune infiltration. Finally, a five-gene (SERPINE1, TNF, CA9, CX3CL1 and JAK3) prognostic model was constructed. Patients in the high-risk group displayed unfavorable survival. Immune infiltration analysis found that the low-risk group had significantly higher levels of immune cell infiltration such as T cells, Macrophages_M1, relative to the high-risk group, and were significantly enriched in apoptosis-associated pathways, which predicted a higher level of immunity. Drug sensitivity correlation analysis revealed that the high-risk group was resistant to conventional chemotherapeutic drugs and sensitive to the Food and Drug Administration-approved drugs BI.2536_1086 and SCH772984_1564. CONCLUSIONS: In the present study, we first found that PCD-related gene expression patterns were correlated with clinical features of CESC patients, which predicts the feasibility of subsequent mining of prognostic features based on these genes. The five-PCD-associated-gene prognostic model showed good assessment ability in predicting patient prognosis, immune response and drug-sensitive response, and provided guidance for the elucidation of the mechanism by which PCD affects CESC, as well as for the clinical targeting of drugs.


Subject(s)
Carcinoma, Squamous Cell , Uterine Cervical Neoplasms , United States , Humans , Female , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics , Prognosis , Apoptosis , Biomarkers , Tumor Microenvironment/genetics
5.
FASEB J ; 34(6): 8544-8557, 2020 06.
Article in English | MEDLINE | ID: mdl-32356314

ABSTRACT

MicroRNAs (miRNAs) play important roles in posttranscriptional regulation and may serve as targets for the diagnosis and treatment of cancers. Nevertheless, a comprehensive understanding of miRNAs profiles in gastric cancer progression is still lacking. Here, we report that miR-129-5p is downregulated in gastric cancer by analyzing TCGA database (n = 41) and clinical tumor samples (n = 60). MiR-129-5p transfection suppressed gastric cancer cell proliferation through inducing G1 phase arrest in vitro and inhibit xenograft tumor growth in vivo. MiR-129-5p directly targeted the 3' untranslated regions (3' UTR) of HOXC10 mRNA and downregulated its expression. Importantly, miR-129-5p could reverse the oncogenic effect induced by HOXC10. We systemically screened the downstream target of HOXC10 by ChIP sequencing, and found that HOXC10 could transcriptionally regulate the expression of Cyclin D1 and facilitate G1/S cell cycle transition. Notably, high levels of HOXC10 and Cyclin D1 were related with poor prognosis of gastric cancer patients (n = 90). These findings reveal a novel role of miR-129-5p/HOXC10/Cyclin D1 axis in modulating cell cycle and gastric tumorigenesis, which might provide potential prognostic biomarkers and therapeutic targets for gastric cancer patients.


Subject(s)
Cell Cycle Checkpoints/genetics , Cyclin D1/genetics , Homeodomain Proteins/genetics , MicroRNAs/genetics , Stomach Neoplasms/genetics , 3' Untranslated Regions/genetics , Animals , Cell Line , Cell Line, Tumor , Cell Proliferation/genetics , Disease Progression , Down-Regulation/genetics , Female , G1 Phase/genetics , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Oncogenes/genetics , S Phase/genetics , Stomach/pathology
6.
PeerJ ; 12: e17861, 2024.
Article in English | MEDLINE | ID: mdl-39119106

ABSTRACT

Background: As a heterogeneous malignancy, breast cancer (BRCA) shows high incidence and mortality. Discovering novel molecular markers and developing reliable prognostic models may improve the survival of BCRA. Methods: The RNA-seq data of BRCA patients were collected from the training set The Cancer Genome Atlas (TCGA)-BRCA and validation set GSE20685 in the Gene Expression Omnibus (GEO) databases. The "GSVA" R package was used to calculate the glycolysis score for each patient, based on which all the patients were divided into different glycolysis groups. The "limma" package was employed to perform differentially expression genes (DEGs) analysis. Key signature genes were selected by performing un/multivariate and least absolute shrinkage and selection operator (LASSO) C regression and used to develop a RiskScore model. The ESTIMATE and MCP-Counter algorithms were used for quantifying immune infiltration level. The functions of the genes were validated using Western blot, colony formation, transwell and wound-healing assay. Results: The glycolysis score and prognostic analysis showed that high glycolysis score was related to tumorigenesis pathway and a poor prognosis in BRCA as overactive glycolysis inhibited the normal functions of immune cells. Subsequently, we screened five key prognostic genes using the LASSO Cox regression analysis and used them to establish a RiskScore with a high classification efficiency. Based on the results of the RiskScore, it was found that patients in the high-risk group had significantly unfavorable immune infiltration and prognostic outcomes. A nomogram integrating the RiskScore could well predict the prognosis for BRCA patients. Knockdown of PSCA suppressed cell proliferation, invasion and migration of BRCA cells. Conclusion: This study developed a glycolysis-related signature with five genes to distinguish between high-risk and low-risk BRCA patients. A nomogram developed on the basis of the RiskScore was reliable to predict BRCA survival. Our model provided clinical guidance for the treatment of BRCA patients.


Subject(s)
Breast Neoplasms , Glycolysis , Humans , Glycolysis/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/mortality , Female , Prognosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Gene Expression Profiling
7.
Sci Rep ; 14(1): 13457, 2024 06 12.
Article in English | MEDLINE | ID: mdl-38862656

ABSTRACT

Acute pancreatitis (AP) is currently among the most prevalent digestive diseases. The pathogenesis of AP remains elusive, and there is no specific treatment. Therefore, identifying novel therapeutic targets is imperative for effective management and prevention of AP. In this study, we conducted a comprehensive transcriptomic analysis of peripheral blood from patients with AP and the pancreatic tissue from a mouse model of AP. Our analyses revealed that mouse model of AP exhibited a higher enrichment of mitogen-activated protein kinase signaling, endocytosis, apoptosis and tight junction pathways than the control. Subsequent weighted gene co-expression network analysis identified 15 gene modules, containing between 50 and 1000 genes each, which demonstrated significant correlations within samples from patients with AP. Further screening identified four genes (ACSL4, GALNT3, WSB1, and IL1R1) that were significantly upregulated in severe acute pancreatitis (SAP) in both human and mouse samples. In mouse models of SAP, ACSL4 was significantly upregulated in the pancreas, whereas GALNT3, WSB1, and IL1R1 were not. Lastly, we found that a commercially available ACSL4 inhibitor, PRGL493, markedly reduced IL-6 and TNFα expression, alleviated pancreatic edema and necrosis, and diminished the infiltration of inflammatory cells. In conclusion, this study comprehensively depicts the key genes and signaling pathways implicated in AP and suggests the potential of ACSL4 as a novel therapeutic target for SAP. These findings provide valuable insights for further exploration of therapeutic strategies for SAP.


Subject(s)
Disease Models, Animal , Pancreatitis , Animals , Pancreatitis/metabolism , Pancreatitis/pathology , Pancreatitis/drug therapy , Pancreatitis/genetics , Humans , Mice , Male , Pancreas/metabolism , Pancreas/pathology , Pancreas/drug effects , Gene Expression Profiling , Signal Transduction , Acute Disease , Female
8.
Sci Adv ; 10(2): eadh9871, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38215194

ABSTRACT

Cell fate transition involves dynamic changes of gene regulatory network and chromatin landscape, requiring multiple levels of regulation, yet the cross-talk between epitranscriptomic modification and chromatin signaling remains largely unknown. Here, we uncover that suppression of N-acetyltransferase 10 (NAT10), the writer for mRNA N4-acetylcytidine (ac4C) modification, can notably affect human embryonic stem cell (hESC) lineage differentiation and pluripotent reprogramming. With integrative analysis, we identify that NAT10-mediated ac4C modification regulates the protein levels of a subset of its targets, which are strongly enriched for fate-instructive chromatin regulators, and among them, histone chaperone ANP32B is experimentally verified and functionally relevant. Furthermore, NAT10-ac4C-ANP32B axis can modulate the chromatin landscape of their downstream genes (e.g., key regulators of Wnt and TGFß pathways). Collectively, we show that NAT10 is an essential regulator of cellular plasticity, and its catalyzed mRNA cytidine acetylation represents a critical layer of epitranscriptomic modulation and uncover a previously unrecognized, direct cross-talk between epitranscriptomic modification and chromatin signaling during cell fate transitions.


Subject(s)
Chromatin , N-Terminal Acetyltransferases , RNA, Messenger , Humans , Acetylation , Acetyltransferases/metabolism , Chromatin/genetics , Cytidine , N-Terminal Acetyltransferases/genetics , N-Terminal Acetyltransferases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Cell Differentiation/genetics
9.
Sci Rep ; 13(1): 17484, 2023 10 14.
Article in English | MEDLINE | ID: mdl-37838783

ABSTRACT

Worldwide, Lung cancer is the leading cause of cancer-related death and poses a direct health threat, non-small cell lung cancer (NSCLC) is the most common type. In this study, we demonstrated that centrosomal protein 20 (CEP20) is upregulated in NSCLC tissues and associated with cancer invasion metastasis. Notably, CEP20 depletion inhibited NSCLC cell proliferation, migration, and microtubule polymerization. Mechanistically, we discovered that CEP20 is critical in the development of NSCLC by regulating microtubule dynamics and cell adhesion-related signaling pathways. Furthermore, the knockdown or overexpression of CEP20 affects microtubule polymerization in A549 cell lines. Our research provides a promising therapeutic target for the diagnosis and treatment of lung cancer, as well as a theoretical and experimental basis for clinical application.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , MicroRNAs , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation , A549 Cells , Transcription Factors/metabolism , Microtubules/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism
10.
Nat Cell Biol ; 25(8): 1146-1156, 2023 08.
Article in English | MEDLINE | ID: mdl-37550515

ABSTRACT

Cellular reprogramming by only small molecules holds enormous potentials for regenerative medicine. However, chemical reprogramming remains a slow process and labour intensive, hindering its broad applications and the investigation of underlying molecular mechanisms. Here, through screening of over 21,000 conditions, we develop a fast chemical reprogramming (FCR) system, which significantly improves the kinetics of cell identity rewiring. We find that FCR rapidly goes through an interesting route for pluripotent reprogramming, uniquely transitioning through a developmentally diapause-like state. Furthermore, FCR critically enables comprehensive characterizations using multi-omics technologies, and has revealed unexpected important features including key regulatory factors and epigenetic dynamics. Particularly, activation of pluripotency-related endogenous retroviruses via inhibition of heterochromatin significantly enhances reprogramming. Our studies provide critical insights into how only environmental cues are sufficient to rapidly reinstate pluripotency in somatic cells, and make notable technical and conceptual advances for solving the puzzle of regeneration.


Subject(s)
Diapause , Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Animals , Cellular Reprogramming/genetics , Cellular Reprogramming Techniques , Regenerative Medicine
11.
Materials (Basel) ; 15(9)2022 May 07.
Article in English | MEDLINE | ID: mdl-35591684

ABSTRACT

Fe-Si-Cr soft magnetic powder cores (SMCs), with high electrical resistivity, magnetic permeability, saturation magnetic induction, and good corrosion resistance, are widely applied to inductors, filters, choke coils, etc. However, with the development of electronic technology with high frequency and high power density, the relative decline in the magnetic properties limits the high-frequency application of SMCs. In this paper, the phosphating process and polyimide (PI) insulation coating is applied to Fe-Si-Cr SMCs to reduce the core loss, including hysteresis loss and eddy current loss. The microstructure and composition of Fe-Si-Cr powders were analyzed by SEM, XRD, and Fourier-transform infrared spectra, respectively. The structural characteristics of the Fe-Si-Cr @ phosphate layer @ PI layer core-shell double coating were studied, and the best process parameters were determined through experiments. For SMCs with 0.4 wt% content of PI, the relative permeability is greater than 68%, and the core loss is the lowest, 7086 mW/cm3; annealed at 500 °C, the relative permeability is greater than 57%, and the core loss is the lowest, 6222 mW/cm3. A 0.4 wt% content of PI, annealed at 500 °C, exhibits the ideal magnetic properties: µe = 47 H/m, P = 6222 mW/cm3.

12.
Ann Transl Med ; 10(21): 1171, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36467343

ABSTRACT

Background: Cervical cancer patients have a high risk of metastasis and a poor prognosis with shorter disease-free survival. Thus, novel biomarkers and feasible therapies urgently need to be discovered. Previous studies have shown that miR-95-3p plays crucial roles in several cancer types. However, the roles of miR-95-3p in cervical cancer remain unknown. Methods: The micro ribonucleic acid (miRNA) expression data and clinical characteristics of cervical cancer samples were downloaded from The Cancer Genome Atlas (TCGA) database. Univariate and multivariate Cox regression analyses were conducted to identify the prognostic-related miRNAs. The potential target genes of miR-95-3p were predicted by the TargetScan database. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were conducted to explore the target gene of miR-95-3p. The effects of miR-95-3p inhibition and overexpression on cell proliferation were inspected by cell counting kit-8 (CCK-8) assays and cell colony formation assays. Wound-healing assays and transwell assays were also used to examine cell migration ability in HeLa and SiHa cells. Results: MiR-95-3p was the only miRNA significantly associated with the poor prognosis of cervical squamous cell carcinoma. A further analysis suggested that vascular cell adhesion molecule 1 (VCAM1) is a target gene of miR-95-3p in cervical cancer, and miR-95-3p promotes the malignant behavior of cervical cancer cells by inhibiting the expression of VCAM1. The CCK-8 and cell colony assays showed that miR-95-3p downregulation significantly suppressed cell proliferation in the HeLa and SiHa cells. The transwell and wound-healing assays showed that miR-95-3p inhibition suppressed cell migration in the HeLa and SiHa cells. Further the Western blot analysis and the quantitative real-time-polymerase chain reaction (qRT-PCR) showed that the knockdown of miR-95-3p in HeLa cells resulted in increased VCAM1 expression. And VCAM1 was highly expressed in the paired adjacent normal cervical epithelium tissue samples, but lowly expressed in the cervical tumor tissue samples. Conclusions: Our study was the first to show that miR-95-3p could serve as a prognostic biomarker of cervical cancer. Mechanistically, we discovered that miR-95-3p inhibited the expression of the cell adhesion molecule VCAM1 and thus promoted further tumor progression.

13.
Nat Commun ; 13(1): 4148, 2022 07 18.
Article in English | MEDLINE | ID: mdl-35851388

ABSTRACT

Pancreatic differentiation from human pluripotent stem cells (hPSCs) provides promising avenues for investigating development and treating diseases. N6-methyladenosine (m6A) is the most prevalent internal messenger RNA (mRNA) modification and plays pivotal roles in regulation of mRNA metabolism, while its functions remain elusive. Here, we profile the dynamic landscapes of m6A transcriptome-wide during pancreatic differentiation. Next, we generate knockout hPSC lines of the major m6A demethylase ALKBH5, and find that ALKBH5 plays significant regulatory roles in pancreatic organogenesis. Mechanistic studies reveal that ALKBH5 deficiency reduces the mRNA stability of key pancreatic transcription factors in an m6A and YTHDF2-dependent manner. We further identify that ALKBH5 cofactor α-ketoglutarate can be applied to enhance differentiation. Collectively, our findings identify ALKBH5 as an essential regulator of pancreatic differentiation and highlight that m6A modification-mediated mRNA metabolism presents an important layer of regulation during cell-fate specification and holds great potentials for translational applications.


Subject(s)
AlkB Homolog 5, RNA Demethylase , RNA Stability , Adenosine/analogs & derivatives , AlkB Homolog 5, RNA Demethylase/metabolism , Humans , Organogenesis/genetics , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/genetics
14.
Sci Adv ; 8(8): eabk1826, 2022 02 25.
Article in English | MEDLINE | ID: mdl-35196077

ABSTRACT

An unlimited source of human pancreatic ß cells is in high demand. Even with recent advances in pancreatic differentiation from human pluripotent stem cells, major hurdles remain in large-scale and cost-effective production of functional ß cells. Here, through chemical screening, we demonstrate that the bromodomain and extraterminal domain (BET) inhibitor I-BET151 can robustly promote the expansion of PDX1+NKX6.1+ pancreatic progenitors (PPs). These expandable PPs (ePPs) maintain pancreatic progenitor cell status in the long term and can efficiently differentiate into functional pancreatic ß (ePP-ß) cells. Notably, transplantation of ePP-ß cells rapidly ameliorated diabetes in mice, suggesting strong potential for cell replacement therapy. Mechanistically, I-BET151 activates Notch signaling and promotes the expression of key PP-associated genes, underscoring the importance of epigenetic and transcriptional modulations for lineage-specific progenitor self-renewal. In summary, our studies achieve the long-term goal of robust expansion of PPs and represent a substantial step toward unlimited supplies of functional ß cells for biomedical research and regenerative medicine.


Subject(s)
Diabetes Mellitus , Insulin-Secreting Cells , Pluripotent Stem Cells , Animals , Cell Differentiation , Diabetes Mellitus/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Trans-Activators/genetics , Trans-Activators/metabolism
15.
Cell Death Dis ; 11(8): 618, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32796817

ABSTRACT

Chemoresistance of tumors often leads to treatment failure in clinical practice, which underscores pivotal needs to uncover novel therapeutic strategies. Accumulating evidences show that microRNAs (miRNAs) are widely involved in carcinogenesis, but their function on chemoresistance remains largely unexplored. In this study, we found that miR-93-5p (miR-93) significantly inhibited cell proliferation, induced G1/S cell cycle arrest and increased chemosensitivity to paclitaxel (PTX) in vitro and in vivo. Moreover, two well-established oncogenes, E2F1 and CCND1, were identified as dual targets of miR-93. Knockdown of E2F1 and CCND1 reduced cell proliferation and PTX-sensitivity, whereas overexpression of them had the opposite effect. More importantly, overexpression of E2F1 and CCND1 antagonized miR-93-mediated cell cycle arrest and apoptosis. Further mechanistic study revealed that miR-93 exhibited its inhibitory role by directly targeting E2F1 and CCND1 to inactivate pRB/E2F1 pathway and AKT phosphorylation. Taken together, our findings suggested that miR-93 greatly improved chemosensitivity and potentially served as a novel therapeutic target for breast cancer treatment.


Subject(s)
Breast Neoplasms/genetics , Carcinogenesis/genetics , Carcinogenesis/pathology , Cyclin D1/metabolism , E2F1 Transcription Factor/metabolism , MicroRNAs/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation/genetics , DNA Methylation/genetics , Down-Regulation/genetics , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Mice, Nude , MicroRNAs/genetics , Minichromosome Maintenance Complex Component 7/genetics , Minichromosome Maintenance Complex Component 7/metabolism , Models, Biological , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-akt/metabolism , Retinoblastoma Protein/metabolism
16.
Cell Death Dis ; 10(11): 807, 2019 10 24.
Article in English | MEDLINE | ID: mdl-31649243

ABSTRACT

Lacking of both prognostic biomarkers and therapeutic targets, triple-negative breast cancer (TNBC) underscores pivotal needs to uncover novel biomarkers and viable therapies. MicroRNAs have broad biological functions in cancers and may serve as ideal biomarkers. In this study, by data mining of the Cancer Genome Atlas database, we screened out 4 differentially-expressed microRNAs (DEmiRNAs) between TNBC and normal samples: miR-135b-5p, miR-9-3p, miR-135b-3p and miR-455-5p. They were specially correlated with the prognosis of TNBC but not non-TNBC. The weighted correlation network analysis (WGCNA) for potential target genes of 3 good prognosis-related DEmiRNAs (miR-135b-5p, miR-9-3p, miR-135b-3p) identified 4 hub genes with highly positive correlation with TNBC subtype: FOXC1, BCL11A, FAM171A1 and RGMA. The targeting relationships between miR-9-3p and FOXC1/FAM171A1, miR-135b-3p and RGMA were validated by dual-luciferase reporter assays. Importantly, the regulatory functions of 4 DEmiRNAs and 3 verified target genes on cell proliferation and migration were explored in TNBC cell lines. In conclusion, we shed lights on these 4 DEmiRNAs (miR-135b-5p, miR-9-3p, miR-135b-3p, miR-455-5p) and 3 hub genes (FOXC1, FAM171A1, RGMA) as specific prognostic biomarkers and promising therapeutic targets for TNBC.


Subject(s)
Biomarkers, Tumor/genetics , Neoplasm Proteins/genetics , Prognosis , Triple Negative Breast Neoplasms/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , Triple Negative Breast Neoplasms/pathology
17.
Cell Death Dis ; 10(9): 628, 2019 08 19.
Article in English | MEDLINE | ID: mdl-31427565

ABSTRACT

Centriole duplication is tightly controlled to occur once per cell cycle, and disruption of this synchrony causes centriole amplification, which is frequently observed in many cancers. Our previous work showed that nuclear distribution gene C (NudC)-like protein 2 (NudCL2) localizes to centrosomes; however, little is known about the role of NudCL2 in the regulation of centrosome function. Here, we find that NudCL2 is required for accurate centriole duplication by stabilizing the E3 ligase HECT domain and RCC1-like domain-containing protein 2 (HERC2). Knockout (KO) of NudCL2 using CRISPR/Cas9-based genome editing or depletion of NudCL2 using small interfering RNA causes significant centriole amplification. Overexpression of NudCL2 significantly suppresses hydroxyurea-induced centriole overduplication. Quantitative proteomic analysis reveals that HERC2 is downregulated in NudCL2 KO cells. NudCL2 is shown to interact with and stabilize HERC2. Depletion of HERC2 leads to the similar defects to that in NudCL2-downregulated cells, and ectopic expression of HERC2 effectively rescues the centriole amplification caused by the loss of NudCL2, whereas the defects induced by HERC2 depletion cannot be reversed by exogenous expression of NudCL2. Either loss of NudCL2 or depletion of HERC2 leads to the accumulation of ubiquitin-specific peptidase 33 (USP33), a centrosomal protein that positively regulates centriole duplication. Moreover, knockdown of USP33 reverses centriole amplification in both NudCL2 KO and HERC2-depleted cells. Taken together, our data suggest that NudCL2 plays an important role in maintaining the fidelity of centriole duplication by stabilizing HERC2 to control USP33 protein levels, providing a previously undescribed mechanism restraining centriole amplification.


Subject(s)
Cell Cycle Proteins/genetics , Centrioles/genetics , Guanine Nucleotide Exchange Factors/genetics , Nuclear Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Centrioles/metabolism , Down-Regulation , Gene Knockout Techniques , Guanine Nucleotide Exchange Factors/biosynthesis , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , HeLa Cells , Humans , Nuclear Proteins/metabolism , Transfection , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL