Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Materials (Basel) ; 15(6)2022 Mar 18.
Article in English | MEDLINE | ID: mdl-35329719

ABSTRACT

Five-axis working machines are applied in the high-precision machining of complex convex surfaces. Therefore, this study integrated a horizontal parallel three-axis motion platform and a three-axis machine tools to create a reconfigurable precision five-axis machine tools (RPFMT). A DELTA OPEN computer numerical control controller was used as the control system architecture. A human-machine interface and programmable controller were incorporated into the developed tool to achieve automatic online measurement. A suitable cutting tool was selected to calculate the five-axis NC machining code for a complex convex surface. The NC codes were input into the LabVIEW software for five-axis postprocessing conversion. A concave workpiece was cut through rough and finishing machining to verify the accuracy of the produced RPFMT.

2.
Cancer Cell ; 39(7): 958-972.e8, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34048709

ABSTRACT

N6-Methyladenosine (m6A) on mRNAs mediates different biological processes and its dysregulation contributes to tumorigenesis. How m6A dictates its diverse molecular and cellular effects in leukemias remains unknown. We found that YTHDC1 is the essential m6A reader in myeloid leukemia from a genome-wide CRISPR screen and that m6A is required for YTHDC1 to undergo liquid-liquid phase separation and form nuclear YTHDC1-m6A condensates (nYACs). The number of nYACs increases in acute myeloid leukemia (AML) cells compared with normal hematopoietic stem and progenitor cells. AML cells require the nYACs to maintain cell survival and the undifferentiated state that is critical for leukemia maintenance. Furthermore, nYACs enable YTHDC1 to protect m6A-mRNAs from the PAXT complex and exosome-associated RNA degradation. Collectively, m6A is required for the formation of a nuclear body mediated by phase separation that maintains mRNA stability and control cancer cell survival and differentiation.


Subject(s)
Adenosine/analogs & derivatives , Cell Nucleus/metabolism , DNA Methylation , Leukemia, Myeloid, Acute/prevention & control , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA Splicing Factors/metabolism , RNA, Messenger/metabolism , Adenosine/chemistry , Adenosine/metabolism , Animals , Apoptosis , Cell Differentiation , Cell Nucleus/genetics , Cell Proliferation , Female , Hematopoiesis , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Liquid-Liquid Extraction , Mice , Mice, Inbred NOD , Mice, SCID , Nerve Tissue Proteins/genetics , Phase Transition , Proto-Oncogene Proteins c-myc/genetics , RNA Splicing Factors/genetics , RNA Stability , RNA, Messenger/chemistry , RNA, Messenger/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Cancer Res ; 81(9): 2470-2480, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33685991

ABSTRACT

PI3Kα inhibitors have shown clinical activity in PIK3CA-mutated estrogen receptor-positive (ER+) patients with breast cancer. Using whole genome CRISPR/Cas9 sgRNA knockout screens, we identified and validated several negative regulators of mTORC1 whose loss confers resistance to PI3Kα inhibition. Among the top candidates were TSC1, TSC2, TBC1D7, AKT1S1, STK11, MARK2, PDE7A, DEPDC5, NPRL2, NPRL3, C12orf66, SZT2, and ITFG2. Loss of these genes invariably results in sustained mTOR signaling under pharmacologic inhibition of the PI3K-AKT pathway. Moreover, resistance could be prevented or overcome by mTOR inhibition, confirming the causative role of sustained mTOR activity in limiting the sensitivity to PI3Kα inhibition. Cumulatively, genomic alterations affecting these genes are identified in about 15% of PIK3CA-mutated breast tumors and appear to be mutually exclusive. This study improves our understanding of the role of mTOR signaling restoration in leading to resistance to PI3Kα inhibition and proposes therapeutic strategies to prevent or revert this resistance. SIGNIFICANCE: These findings show that genetic lesions of multiple negative regulators of mTORC1 could limit the efficacy of PI3Kα inhibitors in breast cancer, which may guide patient selection strategies for future clinical trials.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Drug Resistance, Neoplasm/drug effects , Loss of Function Mutation , Mechanistic Target of Rapamycin Complex 1/metabolism , Phosphoinositide-3 Kinase Inhibitors/administration & dosage , Signal Transduction/genetics , Thiazoles/administration & dosage , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Survival/drug effects , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Cohort Studies , Female , HEK293 Cells , Humans , MCF-7 Cells , Mice , TOR Serine-Threonine Kinases/metabolism , Transduction, Genetic , Tumor Burden/drug effects , Tumor Burden/genetics , Xenograft Model Antitumor Assays
4.
Cell Metab ; 33(1): 145-159.e6, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33357456

ABSTRACT

A significant increase in dietary fructose consumption has been implicated as a potential driver of cancer. Metabolic adaptation of cancer cells to utilize fructose confers advantages for their malignant growth, but compelling therapeutic targets have not been identified. Here, we show that fructose metabolism of leukemic cells can be inhibited by targeting the de novo serine synthesis pathway (SSP). Leukemic cells, unlike their normal counterparts, become significantly dependent on the SSP in fructose-rich conditions as compared to glucose-rich conditions. This metabolic program is mediated by the ratio of redox cofactors, NAD+/NADH, and the increased SSP flux is beneficial for generating alpha-ketoglutarate from glutamine, which allows leukemic cells to proliferate even in the absence of glucose. Inhibition of PHGDH, a rate-limiting enzyme in the SSP, dramatically reduces leukemia engraftment in mice in the presence of high fructose, confirming the essential role of the SSP in the metabolic plasticity of leukemic cells.


Subject(s)
Fructose/metabolism , Leukemia, Myeloid, Acute/metabolism , Serine/biosynthesis , Animals , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Congenic , Mice, Inbred NOD , Tumor Cells, Cultured
5.
Cell Stem Cell ; 27(1): 3-5, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32619516

ABSTRACT

In this issue of Cell Stem Cell, Shen et al. (2020) and Wang et al. (2020) independently identify the essential function of m6A demethylase ALKBH5 in maintaining myeloid leukemia stem cells. These studies expand the regulators of the epitranscriptome that are required for acute myeloid leukemia (AML) development.


Subject(s)
Cell Self Renewal , Leukemia, Myeloid, Acute , AlkB Homolog 5, RNA Demethylase , Carcinogenesis , Humans , Stem Cells
6.
Nat Genet ; 52(2): 198-207, 2020 02.
Article in English | MEDLINE | ID: mdl-31932695

ABSTRACT

Mutations in ARID1A, a subunit of the SWI/SNF chromatin remodeling complex, are the most common alterations of the SWI/SNF complex in estrogen-receptor-positive (ER+) breast cancer. We identify that ARID1A inactivating mutations are present at a high frequency in advanced endocrine-resistant ER+ breast cancer. An epigenome CRISPR-CAS9 knockout (KO) screen identifies ARID1A as the top candidate whose loss determines resistance to the ER degrader fulvestrant. ARID1A inactivation in cells and in patients leads to resistance to ER degraders by facilitating a switch from ER-dependent luminal cells to ER-independent basal-like cells. Cellular plasticity is mediated by loss of ARID1A-dependent SWI/SNF complex targeting to genomic sites of the luminal lineage-determining transcription factors including ER, forkhead box protein A1 (FOXA1) and GATA-binding factor 3 (GATA3). ARID1A also regulates genome-wide ER-FOXA1 chromatin interactions and ER-dependent transcription. Altogether, we uncover a critical role for ARID1A in maintaining luminal cell identity and endocrine therapeutic response in ER+ breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm/genetics , Receptors, Estrogen/metabolism , Transcription Factors/genetics , Animals , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Case-Control Studies , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Female , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Mice , Mutation , Receptors, Estrogen/genetics , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
7.
Cell Rep ; 28(7): 1703-1716.e6, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31412241

ABSTRACT

Stem cells balance cellular fates through asymmetric and symmetric divisions in order to self-renew or to generate downstream progenitors. Symmetric commitment divisions in stem cells are required for rapid regeneration during tissue damage and stress. The control of symmetric commitment remains poorly defined. Using single-cell RNA sequencing (scRNA-seq) in combination with transcriptomic profiling of HSPCs (hematopoietic stem and progenitor cells) from control and m6A methyltransferase Mettl3 conditional knockout mice, we found that m6A-deficient hematopoietic stem cells (HSCs) fail to symmetrically differentiate. Dividing HSCs are expanded and are blocked in an intermediate state that molecularly and functionally resembles multipotent progenitors. Mechanistically, RNA methylation controls Myc mRNA abundance in differentiating HSCs. We identified MYC as a marker for HSC asymmetric and symmetric commitment. Overall, our results indicate that RNA methylation controls symmetric commitment and cell identity of HSCs and may provide a general mechanism for how stem cells regulate differentiation fate choice.


Subject(s)
Cell Differentiation , Cell Lineage , Hematopoiesis , Hematopoietic Stem Cells/cytology , Methyltransferases/physiology , Proto-Oncogene Proteins c-myc/metabolism , Animals , Female , Hematopoietic Stem Cells/metabolism , High-Throughput Nucleotide Sequencing , Male , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-myc/genetics , RNA Stability , Single-Cell Analysis
8.
Cancer Cell ; 36(2): 139-155.e10, 2019 08 12.
Article in English | MEDLINE | ID: mdl-31327655

ABSTRACT

The mechanisms that enable immune evasion at metastatic sites are poorly understood. We show that the Polycomb Repressor Complex 1 (PRC1) drives colonization of the bones and visceral organs in double-negative prostate cancer (DNPC). In vivo genetic screening identifies CCL2 as the top prometastatic gene induced by PRC1. CCL2 governs self-renewal and induces the recruitment of M2-like tumor-associated macrophages and regulatory T cells, thus coordinating metastasis initiation with immune suppression and neoangiogenesis. A catalytic inhibitor of PRC1 cooperates with immune checkpoint therapy to reverse these processes and suppress metastasis in genetically engineered mouse transplantation models of DNPC. These results reveal that PRC1 coordinates stemness with immune evasion and neoangiogenesis and point to the potential clinical utility of targeting PRC1 in DNPC.


Subject(s)
Adenocarcinoma/metabolism , Cell Movement , Cell Self Renewal , Chemokine CCL2/metabolism , Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 1/metabolism , Prostatic Neoplasms/metabolism , Tumor Escape , Adenocarcinoma/drug therapy , Adenocarcinoma/immunology , Adenocarcinoma/secondary , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Movement/drug effects , Cell Self Renewal/drug effects , Chemokine CCL2/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, Nude , Mice, SCID , Neoplasm Metastasis , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , PC-3 Cells , Polycomb Repressive Complex 1/antagonists & inhibitors , Polycomb Repressive Complex 1/genetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Receptors, Androgen/deficiency , Receptors, Androgen/genetics , Receptors, CCR4/genetics , Receptors, CCR4/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tumor Escape/drug effects , Xenograft Model Antitumor Assays
9.
Cancer Discov ; 9(1): 25-33, 2019 01.
Article in English | MEDLINE | ID: mdl-30578356

ABSTRACT

Hematopoietic development and differentiation are highly regulated processes, and recent studies focusing on m6A mRNA methylation have uncovered how this mark controls cell fate in both normal and malignant hematopoietic states. In this review, we focus on how writers, readers, and erasers of RNA methylation can mediate distinct phenotypes on mRNAs and on cells. Targeting the RNA methylation program has emerged as a potential novel therapeutic strategy, and we explore the role for these regulators in both normal and dysregulated cell contexts. SIGNIFICANCE: RNA methylation is required for cancer cell survival in solid tumors and in acute myeloid leukemia, and targeting this pathway has been proposed as a new therapeutic strategy in cancer. However, understanding the role for RNA methylation in both normal and malignant states is essential for understanding the potential consequences for therapeutic intervention.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , RNA, Messenger/metabolism , Animals , Cell Differentiation , Gene Expression Regulation, Neoplastic , Hematopoiesis , Humans , Leukemia, Myeloid, Acute/genetics , Methylation , RNA, Messenger/genetics
10.
Brief Bioinform ; 19(5): 905-917, 2018 09 28.
Article in English | MEDLINE | ID: mdl-28387786

ABSTRACT

RNA-sequencing (RNA-seq) can generate millions of reads to provide clues for analyzing novel or abnormal alternative splicing (AS) events in cells. However, current methods for exploring AS events are still far from being satisfactory. Here, we present Comprehensive AS Hunting (CASH), which constructs comprehensive splice sites including known and novel AS sites in cells, and identifies differentially AS events between cells. We illuminated the versatility of CASH on RNA-seq data from a wide range of species and also on simulated in silico data, validated the advantages of CASH over other AS predictors and exhibited novel differentially AS events. Moreover, we used CASH to identify SRSF10-regulated AS events and investigated the evolution of SRSF10-regulated splicing. The results showed that SRSF10-regulated splicing events are highly evolvable from chickens, mice to humans. However, SRSF10-regulated splicing model was observed to be immutable, in which SRSF10 binding to cassette exon promotes exon inclusion while binding to downstream exon induces exon skipping. Altogether, CASH can significantly improve the detection of AS events and facilitate the study of AS regulation and function in cells; the SRSF10 data first demonstrate a flexibility of SRSF10 with their regulated splicing events but an immutability of SRSF10-regulated splicing model to produce opposite AS outcomes in vertebrates.


Subject(s)
Alternative Splicing , RNA Splice Sites , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chickens , Computational Biology/methods , Computer Simulation , Exons , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/statistics & numerical data , Humans , Mice , Models, Genetic , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sequence Analysis, RNA/methods , Sequence Analysis, RNA/statistics & numerical data , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism , Software
11.
Nat Med ; 23(11): 1369-1376, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28920958

ABSTRACT

N6-methyladenosine (m6A) is an abundant nucleotide modification in mRNA that is required for the differentiation of mouse embryonic stem cells. However, it remains unknown whether the m6A modification controls the differentiation of normal and/or malignant myeloid hematopoietic cells. Here we show that shRNA-mediated depletion of the m6A-forming enzyme METTL3 in human hematopoietic stem/progenitor cells (HSPCs) promotes cell differentiation, coupled with reduced cell proliferation. Conversely, overexpression of wild-type METTL3, but not of a catalytically inactive form of METTL3, inhibits cell differentiation and increases cell growth. METTL3 mRNA and protein are expressed more abundantly in acute myeloid leukemia (AML) cells than in healthy HSPCs or other types of tumor cells. Furthermore, METTL3 depletion in human myeloid leukemia cell lines induces cell differentiation and apoptosis and delays leukemia progression in recipient mice in vivo. Single-nucleotide-resolution mapping of m6A coupled with ribosome profiling reveals that m6A promotes the translation of c-MYC, BCL2 and PTEN mRNAs in the human acute myeloid leukemia MOLM-13 cell line. Moreover, loss of METTL3 leads to increased levels of phosphorylated AKT, which contributes to the differentiation-promoting effects of METTL3 depletion. Overall, these results provide a rationale for the therapeutic targeting of METTL3 in myeloid leukemia.


Subject(s)
Adenosine/analogs & derivatives , Bone Marrow Cells/cytology , Cell Differentiation/physiology , Leukemia, Myeloid, Acute/pathology , Methyltransferases/physiology , Adenosine/biosynthesis , Cells, Cultured , Clustered Regularly Interspaced Short Palindromic Repeats , Humans , Tumor Cells, Cultured
12.
Cancer Res ; 77(5): 1168-1178, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28082404

ABSTRACT

Aberrant RNA splicing is recognized to contribute to cancer pathogenesis, but the underlying mechanisms remain mainly obscure. Here, we report that the splicing factor SRSF2 is upregulated frequently in human hepatocellular carcinoma (HCC), where this event is associated with poor prognosis in patients. RNA-seq and other molecular analyses were used to identify SRSF2-regulated alternative splicing events. SRSF2 binding within an alternative exon was associated with its inclusion in the RNA, whereas SRSF2 binding in a flanking constitutive exon was associated with exclusion of the alternative exon. Notably, cancer-associated splice variants upregulated by SRSF2 in clinical specimens of HCC were found to be crucial for pathogenesis and progression in hepatoma cells, where SRSF2 expression increased cell proliferation and tumorigenic potential by controlling expression of these variants. Our findings identify SRSF2 as a key regulator of RNA splicing dysregulation in cancer, with possible clinical implications as a candidate prognostic factor in patients with HCC. Cancer Res; 77(5); 1168-78. ©2017 AACR.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Serine-Arginine Splicing Factors/genetics , Alternative Splicing , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , HEK293 Cells , Humans , Liver Neoplasms/pathology , Transfection
13.
Mol Cell Biol ; 36(11): 1628-38, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27022105

ABSTRACT

The liver performs a variety of unique functions critical for metabolic homeostasis. Here, we show that mice lacking the splicing factor SRSF2 but not SRSF1 in hepatocytes have severe liver pathology and biochemical abnormalities. Histological analyses revealed generalized hepatitis with the presence of ballooned hepatocytes and evidence of fibrosis. Molecular analysis demonstrated that SRSF2 governs splicing of multiple genes involved in the stress-induced cell death pathway in the liver. More importantly, SRSF2 also functions as a potent transcription activator, required for efficient expression of transcription factors mainly responsible for energy homeostasis and bile acid metabolism in the liver. Consistent with the effects of SRSF2 in gene regulation, accumulation of total cholesterol and bile acids was prominently observed in the mutant liver, followed by enhanced generation of reactive oxygen species and increased endoplasmic reticulum stress, as revealed by biochemical and ultrastructural analyses. Taking these observations together, inactivation of SRSF2 in liver caused dysregulated splicing events and hepatic metabolic disorders, which trigger endoplasmic reticulum stress, oxidative stress, and finally liver failure.


Subject(s)
Hepatocytes/metabolism , Liver Failure, Acute/mortality , Sequence Analysis, RNA/methods , Serine-Arginine Splicing Factors/deficiency , Transcription Factors/genetics , Animals , Bile Acids and Salts/metabolism , Cells, Cultured , Disease Models, Animal , Endoplasmic Reticulum Stress , Energy Metabolism , Gene Expression Profiling/methods , Gene Expression Regulation , Gene Knockout Techniques , Liver Failure, Acute/genetics , Mice , Oxidative Stress , RNA Splicing , Transcriptional Activation
14.
Cell Rep ; 13(8): 1647-57, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26586428

ABSTRACT

Alternative splicing is a major mechanism of controlling gene expression and protein diversity in higher eukaryotes. We report that the splicing factor SRSF10 functions during striated muscle development, myoblast differentiation, and glucose production both in cells and in mice. A combination of RNA-sequencing and molecular analysis allowed us to identify muscle-specific splicing events controlled by SRSF10 that are critically involved in striated muscle development. Inclusion of alternative exons 16 and 17 of Lrrfip1 is a muscle-specific event that is activated by SRSF10 and essential for myoblast differentiation. On the other hand, in mouse primary hepatocytes, PGC1α is a key target of SRSF10 that regulates glucose production by fasting. SRSF10 represses inclusion of PGC1α exon 7a and facilitates the production of functional protein. The results highlight the biological significance of SRSF10 and regulated alternative splicing in vivo.


Subject(s)
Alternative Splicing/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation/genetics , Glucose/metabolism , Myoblasts/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Animals , Cells, Cultured , Exons/genetics , Male , Mice , Mice, Inbred C57BL , Muscle Development/genetics , RNA/genetics , RNA Splicing/genetics , Sequence Analysis, RNA/methods , Serine-Arginine Splicing Factors
15.
Nat Commun ; 5: 4581, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-25091051

ABSTRACT

Bcl-2-associated transcription factor 1 (BCLAF1) is known to be involved in multiple biological processes. Although several splice variants of BCLAF1 have been identified, little is known about how BCLAF1 splicing is regulated or the contribution of alternative splicing to its developmental functions. Here we find that inclusion of alternative exon5a was significantly increased in colorectal cancer (CRC) samples. Knockdown of the BCLAF1 protein isoform resulting from exon5a inclusion inhibited growth and that its overexpression increased tumorigenic potential. We also found that the splicing factor SRSF10 stimulates inclusion of exon5a and has growth-inducing activity. Importantly, the upregulation of SRSF10 expression observed in clinical CRC samples parallels the increased inclusion of BCLAF1 exon5a, both of which are associated with higher tumour grade. These findings identify SRSF10 as a key regulator of BCLAF1 pre-mRNA splicing and the maintenance of oncogenic features in human colon cancer cells.


Subject(s)
Alternative Splicing , Carcinoma/genetics , Cell Cycle Proteins/metabolism , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Antineoplastic Agents/chemistry , Carcinoma/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/metabolism , Exons , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Nude , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , RNA Precursors/genetics , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , Serine-Arginine Splicing Factors , Tumor Suppressor Proteins/genetics , Up-Regulation
16.
Mol Cell Biol ; 34(12): 2198-207, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24710272

ABSTRACT

During adipocyte differentiation, significant alternative splicing changes occur in association with the adipogenic process. However, little is known about roles played by splicing factors in this process. We observed that mice deficient for the splicing factor SRSF10 exhibit severely impaired development of subcutaneous white adipose tissue (WAT) as a result of defects in adipogenic differentiation. To identify splicing events responsible for this, transcriptome sequencing (RNA-seq) analysis was performed using embryonic fibroblast cells. Several SRSF10-affected splicing events that are implicated in adipogenesis have been identified. Notably, lipin1, known as an important regulator during adipogenesis, was further investigated. While lipin1ß is mainly involved in lipogenesis, its alternatively spliced isoform lipin1α, generated through the skipping of exon 7, is primarily required for initial adipocyte differentiation. Skipping of exon 7 is controlled by an SRSF10-regulated cis element located in the constitutive exon 8. The activity of this element depends on the binding of SRSF10 and correlates with the relative abundance of lipin1α mRNA. A series of experiments demonstrated that SRSF10 controls the production of lipin1α and thus promotes adipocyte differentiation. Indeed, lipin1α expression could rescue SRSF10-mediated adipogenic defects. Taken together, our results identify SRSF10 as an essential regulator for adipocyte differentiation and also provide new insights into splicing control by SRSF10 in lipin1 pre-mRNA splicing.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Alternative Splicing/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation/genetics , Neoplasm Proteins/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Adipocytes/drug effects , Adipogenesis/drug effects , Adipogenesis/genetics , Alternative Splicing/drug effects , Animals , Base Sequence , Cell Differentiation/drug effects , Computational Biology , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Exons/genetics , Fibroblasts/drug effects , Fibroblasts/metabolism , Hormones/pharmacology , Mice , Mice, Knockout , Nuclear Proteins/genetics , Phosphatidate Phosphatase/genetics , Protein Binding/drug effects , Protein Binding/genetics , Reproducibility of Results , Sequence Analysis, RNA , Subcutaneous Fat/cytology
17.
Nucleic Acids Res ; 42(6): 4019-30, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24442672

ABSTRACT

Splicing factor SRSF10 is known to function as a sequence-specific splicing activator. Here, we used RNA-seq coupled with bioinformatics analysis to identify the extensive splicing network regulated by SRSF10 in chicken cells. We found that SRSF10 promoted both exon inclusion and exclusion. Motif analysis revealed that SRSF10 binding to cassette exons was associated with exon inclusion, whereas the binding of SRSF10 within downstream constitutive exons was associated with exon exclusion. This positional effect was further demonstrated by the mutagenesis of potential SRSF10 binding motifs in two minigene constructs. Functionally, many of SRSF10-verified alternative exons are linked to pathways of stress and apoptosis. Consistent with this observation, cells depleted of SRSF10 expression were far more susceptible to endoplasmic reticulum stress-induced apoptosis than control cells. Importantly, reconstituted SRSF10 in knockout cells recovered wild-type splicing patterns and considerably rescued the stress-related defects. Together, our results provide mechanistic insight into SRSF10-regulated alternative splicing events in vivo and demonstrate that SRSF10 plays a crucial role in cell survival under stress conditions.


Subject(s)
Alternative Splicing , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Animals , Apoptosis , Cell Line , Chickens , Endoplasmic Reticulum Stress , Exons , Gene Expression Profiling , Nuclear Proteins/genetics , Nucleotide Motifs , RNA/chemistry , RNA/metabolism , RNA-Binding Proteins/genetics , Sequence Analysis, RNA , Serine-Arginine Splicing Factors
18.
Genom Data ; 2: 20-3, 2014 Dec.
Article in English | MEDLINE | ID: mdl-26484059

ABSTRACT

Splicing factor SRSF10 is known to function as a sequence-specific splicing activator that is capable of regulating alternative splicing both in vitro and in vivo. We recently used an RNA-seq approach coupled with bioinformatics analysis to identify the extensive splicing network regulated by SRSF10 in chicken cells. We found that SRSF10 promoted both exon inclusion and exclusion. Functionally, many of the SRSF10-verified alternative exons are linked to pathways of response to external stimulus. Here we describe in detail the experimental design, bioinformatics analysis and GO/pathway enrichment analysis of SRSF10-regulated genes to correspond with our data in the Gene Expression Omnibus with accession number GSE53354. Our data thus provide a resource for studying regulation of alternative splicing in vivo that underlines biological functions of splicing regulatory proteins in cells.

19.
Proc Natl Acad Sci U S A ; 110(29): E2687-95, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23818605

ABSTRACT

Splicing of mRNA precursors consists of two steps that are almost invariably tightly coupled to facilitate efficient generation of spliced mRNA. However, we described previously a splicing substrate that is completely blocked after the first step. We have now investigated the basis for this unusual second-step inhibition and unexpectedly elucidated two independent mechanisms. One involves a stem-loop structure located downstream of the 3'splice site, and the other involves an exonic splicing silencer (ESS) situated 3' to the structure. Both elements contribute to the second-step block in vitro and also cause exon skipping in vivo. Importantly, we identified far upstream element-binding protein 1 (FUBP1), a single-stranded DNA- and RNA-binding protein not previously implicated in splicing, as a strong ESS binding protein, and several assays implicate it in ESS function. We demonstrate using depletion/add-back experiments that FUBP1 acts as a second-step repressor in vitro and show by siRNA-mediated knockdown and overexpression assays that it modulates exon inclusion in vivo. Together, our results provide additional insights into splicing control, and identify FUBP1 as a splicing regulator.


Subject(s)
DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation/physiology , Nucleic Acid Conformation , RNA Splicing/physiology , RNA/metabolism , Base Sequence , Binding Sites/genetics , Blotting, Western , DNA Primers/genetics , Electrophoretic Mobility Shift Assay , Exons/genetics , HeLa Cells , Humans , Molecular Sequence Data , RNA/chemistry , RNA Interference , RNA-Binding Proteins , Reverse Transcriptase Polymerase Chain Reaction
20.
Nan Fang Yi Ke Da Xue Xue Bao ; 30(7): 1584-6, 2010 Jul.
Article in Chinese | MEDLINE | ID: mdl-20650772

ABSTRACT

OBJECTIVE: To evaluate the effect of placement of short iliac screw using Galveston technique in lumbosacral fusion. METHODS: From October 2003 to August 2007, 18 consecutive patients (mean age 46 years ranging from 25 to 62 years) received placement of short iliac screw in lumbosacral fusion. The patients were followed up for a mean of 18 months (12-23 months), and the effect of lumbosacral fusion was evaluated according to standing anterior-posterior and lateral plain films taken before and after the operation and at the follow-up and also on the basis of symptom relief. RESULTS: The mean time of surgery was 210 min (180-290 min). No complications occurred during and after the operation. According to the evaluation criteria of surgical treatment of low back pain formulated by the spine group of Chinese Orthopedic Association, excellent clinical outcome was achieved in 12 cases, good outcome in 3 cases, and tolerable outcome in 2 cases, with the excellent and good outcome rate of 83%. CONCLUSION: The Galveston technique for short iliac screw placement can obtain satisfactory outcome in the lumbosacral fusion.


Subject(s)
Fracture Fixation, Internal/methods , Ilium/surgery , Spinal Fusion/methods , Adult , Bone Screws , Female , Humans , Internal Fixators , Lumbar Vertebrae/surgery , Male , Middle Aged , Sacrum/surgery , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...