Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Mol Cell ; 84(6): 1062-1077.e9, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38309276

ABSTRACT

Inverted Alu repeats (IRAlus) are abundantly found in the transcriptome, especially in introns and 3' untranslated regions (UTRs). Yet, the biological significance of IRAlus embedded in 3' UTRs remains largely unknown. Here, we find that 3' UTR IRAlus silences genes involved in essential signaling pathways. We utilize J2 antibody to directly capture and map the double-stranded RNA structure of 3' UTR IRAlus in the transcriptome. Bioinformatic analysis reveals alternative polyadenylation as a major axis of IRAlus-mediated gene regulation. Notably, the expression of mouse double minute 2 (MDM2), an inhibitor of p53, is upregulated by the exclusion of IRAlus during UTR shortening, which is exploited to silence p53 during tumorigenesis. Moreover, the transcriptome-wide UTR lengthening in neural progenitor cells results in the global downregulation of genes associated with neurodegenerative diseases, including amyotrophic lateral sclerosis, via IRAlus inclusion. Our study establishes the functional landscape of 3' UTR IRAlus and its role in human pathophysiology.


Subject(s)
Polyadenylation , Tumor Suppressor Protein p53 , Humans , Mice , Animals , Tumor Suppressor Protein p53/genetics , 3' Untranslated Regions/genetics , Gene Expression Regulation , Introns
2.
Nature ; 617(7961): 540-547, 2023 May.
Article in English | MEDLINE | ID: mdl-37165195

ABSTRACT

Throughout an individual's lifetime, genomic alterations accumulate in somatic cells1-11. However, the mutational landscape induced by retrotransposition of long interspersed nuclear element-1 (L1), a widespread mobile element in the human genome12-14, is poorly understood in normal cells. Here we explored the whole-genome sequences of 899 single-cell clones established from three different cell types collected from 28 individuals. We identified 1,708 somatic L1 retrotransposition events that were enriched in colorectal epithelium and showed a positive relationship with age. Fingerprinting of source elements showed 34 retrotransposition-competent L1s. Multidimensional analysis demonstrated that (1) somatic L1 retrotranspositions occur from early embryogenesis at a substantial rate, (2) epigenetic on/off of a source element is preferentially determined in the early organogenesis stage, (3) retrotransposition-competent L1s with a lower population allele frequency have higher retrotransposition activity and (4) only a small fraction of L1 transcripts in the cytoplasm are finally retrotransposed in somatic cells. Analysis of matched cancers further suggested that somatic L1 retrotransposition rate is substantially increased during colorectal tumourigenesis. In summary, this study illustrates L1 retrotransposition-induced somatic mosaicism in normal cells and provides insights into the genomic and epigenomic regulation of transposable elements over the human lifetime.


Subject(s)
Colon , DNA Transposable Elements , Intestinal Mucosa , Retroelements , Humans , Carcinogenesis/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Transposable Elements/genetics , Genomics , Long Interspersed Nucleotide Elements/genetics , Retroelements/genetics , Aging/genetics , Gene Frequency , Mosaicism , Epigenomics , Genome, Human/genetics , Colon/metabolism , Intestinal Mucosa/metabolism , Embryonic Development/genetics
3.
Circ Res ; 131(10): 792-806, 2022 10 28.
Article in English | MEDLINE | ID: mdl-36205124

ABSTRACT

BACKGROUND: In large-scale genomic studies, Sox17, an endothelial-specific transcription factor, has been suggested as a putative causal gene of pulmonary arterial hypertension (PAH); however, its role and molecular mechanisms remain to be elucidated. We investigated the functional impacts and acting mechanisms of impaired Sox17 (SRY-related HMG-box17) pathway in PAH and explored its potential as a therapeutic target. METHODS: In adult mice, Sox17 deletion in pulmonary endothelial cells (ECs) induced PAH under hypoxia with high penetrance and severity, but not under normoxia. RESULTS: Key features of PAH, such as hypermuscularization, EC hyperplasia, and inflammation in lung arterioles, right ventricular hypertrophy, and elevated pulmonary arterial pressure, persisted even after long rest in normoxia. Mechanistically, transcriptomic profiling predicted that the combination of Sox17 deficiency and hypoxia activated c-Met signaling in lung ECs. HGF (hepatocyte grow factor), a ligand of c-Met, was upregulated in Sox17-deficient lung ECs. Pharmacologic inhibition of HGF/c-Met signaling attenuated and reversed the features of PAH in both preventive and therapeutic settings. Similar to findings in animal models, Sox17 levels in lung ECs were repressed in 26.7% of PAH patients (4 of 15), while those were robust in all 14 non-PAH controls. HGF levels in pulmonary arterioles were increased in 86.7% of patients with PAH (13 of 15), but none of the controls showed that pattern. CONCLUSIONS: The downregulation of Sox17 levels in pulmonary arterioles increases the susceptibility to PAH, particularly when exposed to hypoxia. Our findings suggest the reactive upregulation of HGF/c-Met signaling as a novel druggable target for PAH treatment.


Subject(s)
Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Animals , Mice , Endothelial Cells/metabolism , HMGB Proteins/metabolism , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypoxia/complications , Hypoxia/metabolism , Pulmonary Arterial Hypertension/genetics , Pulmonary Artery/metabolism , Signal Transduction , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Proto-Oncogene Proteins c-met/metabolism
4.
Cell ; 138(4): 696-708, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19703396

ABSTRACT

As key regulators in cellular functions, microRNAs (miRNAs) themselves need to be tightly controlled. Lin28, a pluripotency factor, was reported to downregulate let-7 miRNA by inducing uridylation of let-7 precursor (pre-let-7). But the enzyme responsible for the uridylation remained unknown. Here we identify a noncanonical poly (A) polymerase, TUTase4 (TUT4), as the uridylyl transferase for pre-let-7. Lin28 recruits TUT4 to pre-let-7 by recognizing a tetra-nucleotide sequence motif (GGAG) in the terminal loop. TUT4 in turn adds an oligouridine tail to the pre-let-7, which blocks Dicer processing. Other miRNAs with the same sequence motif (miR-107, -143, and -200c) are regulated through the same mechanism. Knockdown of TUT4 and Lin28 reduces the level of stem cell markers, suggesting that they are required for stem cell maintenance. This study uncovers the role of TUT4 and Lin28 as specific suppressors of miRNA biogenesis, which has implications for stem cell research and cancer biology.


Subject(s)
Embryonic Stem Cells/cytology , MicroRNAs/metabolism , Polynucleotide Adenylyltransferase/metabolism , Uridine/metabolism , Animals , Cell Line , Gene Knockdown Techniques , Humans , Mice
5.
Cell ; 136(1): 75-84, 2009 Jan 09.
Article in English | MEDLINE | ID: mdl-19135890

ABSTRACT

The Drosha-DGCR8 complex, also known as Microprocessor, is essential for microRNA (miRNA) maturation. Drosha functions as the catalytic subunit, while DGCR8 (also known as Pasha) recognizes the RNA substrate. Although the action mechanism of this complex has been intensively studied, it remains unclear how Drosha and DGCR8 are regulated and if these proteins have any additional role(s) apart from miRNA processing. Here, we report that Drosha and DGCR8 regulate each other posttranscriptionally. The Drosha-DGCR8 complex cleaves the hairpin structures embedded in the DGCR8 mRNA and thereby destabilizes the mRNA. We further find that DGCR8 stabilizes the Drosha protein via protein-protein interaction. This crossregulation between Drosha and DGCR8 may contribute to the homeostatic control of miRNA biogenesis. Furthermore, microarray analyses suggest that a number of mRNAs may be downregulated in a Microprocessor-dependent, miRNA-independent manner. Our study reveals a previously unsuspected function of Microprocessor in mRNA stability control.


Subject(s)
Gene Expression Regulation , Proteins/genetics , RNA Stability , Ribonuclease III/genetics , Animals , Base Sequence , Cell Line , Humans , Molecular Sequence Data , Nucleic Acid Conformation , Proteins/metabolism , RNA Interference , RNA-Binding Proteins , Ribonuclease III/metabolism
6.
Int J Mol Sci ; 23(22)2022 Nov 10.
Article in English | MEDLINE | ID: mdl-36430334

ABSTRACT

Noonan syndrome (NS) is a genetic disorder mainly caused by gain-of-function mutations in Src homology region 2-containing protein tyrosine phosphatase 2 (SHP2). Although diverse neurological manifestations are commonly diagnosed in NS patients, the mechanisms as to how SHP2 mutations induce the neurodevelopmental defects associated with NS remain elusive. Here, we report that cortical organoids (NS-COs) derived from NS-induced pluripotent stem cells (iPSCs) exhibit developmental abnormalities, especially in excitatory neurons (ENs). Although NS-COs develop normally in their appearance, single-cell transcriptomic analysis revealed an increase in the EN population and overexpression of cortical layer markers in NS-COs. Surprisingly, the EN subpopulation co-expressing the upper layer marker SATB2 and the deep layer maker CTIP2 was enriched in NS-COs during cortical development. In parallel with the developmental disruptions, NS-COs also exhibited reduced synaptic connectivity. Collectively, our findings suggest that perturbed cortical layer identity and impeded neuronal connectivity contribute to the neurological manifestations of NS.


Subject(s)
Induced Pluripotent Stem Cells , Noonan Syndrome , Humans , Organoids , Noonan Syndrome/genetics , Brain , Neurons
7.
Nat Rev Mol Cell Biol ; 10(2): 126-39, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19165215

ABSTRACT

Small RNAs of 20-30 nucleotides can target both chromatin and transcripts, and thereby keep both the genome and the transcriptome under extensive surveillance. Recent progress in high-throughput sequencing has uncovered an astounding landscape of small RNAs in eukaryotic cells. Various small RNAs of distinctive characteristics have been found and can be classified into three classes based on their biogenesis mechanism and the type of Argonaute protein that they are associated with: microRNAs (miRNAs), endogenous small interfering RNAs (endo-siRNAs or esiRNAs) and Piwi-interacting RNAs (piRNAs). This Review summarizes our current knowledge of how these intriguing molecules are generated in animal cells.


Subject(s)
MicroRNAs/metabolism , RNA, Small Interfering/metabolism , Animals , Eukaryotic Initiation Factors/chemistry , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Gene Expression Regulation , Humans , MicroRNAs/genetics , Models, Molecular , Protein Conformation , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , Ribonuclease III/chemistry , Ribonuclease III/genetics , Ribonuclease III/metabolism
8.
Genes Dev ; 26(1): 6-10, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22215805

ABSTRACT

Differentiation of multipotent stem cells occurs through the highly coordinated control of gene expression. Repressor element 1 (RE1) silencing transcription factor (REST), a master transcriptional regulator in neuronal stem cells, restricts neuronal gene expression. REST activity is context-dependent and is modified by its cofactors, such as Ctdsp2. In this issue of Genes & Development, Dill and colleagues (pp. 25-30) report on the microRNA-mediated regulation of neural differentiation. Interestingly, this microRNA is post-transcriptionally regulated and modulates expression of its host gene, ctdsp2.


Subject(s)
Cell Differentiation , Gene Expression Regulation, Developmental , MicroRNAs/genetics , MicroRNAs/metabolism , Neurons/cytology , Neurons/metabolism , Phosphoprotein Phosphatases/genetics , Zebrafish Proteins/genetics , Animals
9.
J Neurosci ; 35(12): 4983-98, 2015 Mar 25.
Article in English | MEDLINE | ID: mdl-25810528

ABSTRACT

In the mammalian hippocampus, canonical Wnt signals provided by the microenvironment regulate the differentiation of adult neural stem cells (NSCs) toward the neuronal lineage. Wnts are part of a complex and diverse set of signaling pathways and the role of Wnt/Planar cell polarity (PCP) signaling in adult neurogenesis remains unknown. Using in vitro assays on differentiating adult NSCs, we identified a transition of Wnt signaling responsiveness from Wnt/ß-catenin to Wnt/PCP signaling. In mice, retroviral knockdown strategies against ATP6AP2, a recently discovered core protein involved in both signaling pathways, revealed that its dual role is critical for granule cell fate and morphogenesis. We were able to confirm its dual role in neurogenic Wnt signaling in vitro for both canonical Wnt signaling in proliferating adult NSCs and non-canonical Wnt signaling in differentiating neuroblasts. Although LRP6 appeared to be critical for granule cell fate determination, in vivo knockdown of PCP core proteins FZD3 and CELSR1-3 revealed severe maturational defects without changing the identity of newborn granule cells. Furthermore, we found that CELSR1-3 control distinctive aspects of PCP-mediated granule cell morphogenesis with CELSR1 regulating the direction of dendrite initiation sites and CELSR2/3 controlling radial migration and dendritic patterning. The data presented here characterize distinctive roles for Wnt/ß-catenin signaling in granule cell fate determination and for Wnt/PCP signaling in controlling the morphological maturation of differentiating neuroblasts.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Hippocampus/cytology , Neurogenesis/physiology , Proton-Translocating ATPases/physiology , Receptors, Cell Surface/physiology , Animals , Cadherins/genetics , Cadherins/physiology , Cell Differentiation/physiology , Cell Polarity/physiology , Cells, Cultured , Female , Frizzled Receptors/genetics , Frizzled Receptors/physiology , Gene Expression Regulation, Developmental/genetics , Gene Knockdown Techniques , Hippocampus/growth & development , Mice , Neural Stem Cells/physiology , Neurogenesis/genetics , Proton-Translocating ATPases/genetics , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Signal Transduction/physiology , Up-Regulation , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology , beta Catenin/physiology
10.
Mol Cell ; 32(2): 276-84, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18951094

ABSTRACT

The precise control of microRNA (miRNA) biogenesis is critical for embryonic development and normal cellular functions, and its dysregulation is often associated with human diseases. Though the birth and maturation pathway of miRNA has been established, the regulation and death pathway remains largely unknown. Here, we report the RNA-binding proteins, Lin28a and Lin28b, as posttranscriptional repressors of let-7 miRNA biogenesis. We observe that the Lin28 proteins act mainly in the cytoplasm by inducing uridylation of precursor let-7 (pre-let-7) at its 3' end. The uridylated pre-let-7 (up-let-7) fails Dicer processing and undergoes degradation. We provide a mechanism for the posttranscriptional regulation of miRNA biogenesis by Lin28 which is highly expressed in undifferentiated cells and certain cancer cells. The Lin28-mediated downregulation of let-7 may play a key role in development, stem cell programming, and tumorigenesis.


Subject(s)
DNA-Binding Proteins/physiology , MicroRNAs/metabolism , RNA Precursors/metabolism , RNA-Binding Proteins/physiology , Animals , Cell Line , Cytoplasm/metabolism , DNA-Binding Proteins/genetics , Humans , Mice , MicroRNAs/biosynthesis , Models, Genetic , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/genetics , Ribonuclease III/metabolism
11.
PLoS One ; 19(4): e0301622, 2024.
Article in English | MEDLINE | ID: mdl-38630695

ABSTRACT

This paper proposes a reinforced concrete (RC) boundary beam-wall system that requires less construction material and a smaller floor height compared to the conventional RC transfer girder system. The structural performance of this system subjected to axial compression was evaluated by performing a structural test on four specimens of 1/2 scale. In addition, three-dimensional nonlinear finite element analysis was also performed to verify the effectiveness of the boundary beam-wall system. Three test parameters such as the lower wall length-to-upper wall length ratio, lower wall thickness, and stirrup details of the lower wall were considered. The load-displacement curve was plotted for each specimen and its failure mode was identified. The test results showed that decrease in the lower wall length-to-upper wall length ratio significantly reduced the peak strength of the boundary beam-wall system and difference in upper and lower wall thicknesses resulted in lateral bending caused by eccentricity in the out-of-plane direction. Additionally, incorporating cross-ties and reducing stirrup spacing in the lower wall significantly improved initial stiffness and peak strength, effectively minimizing stress concentration.


Subject(s)
Construction Materials , Data Compression , Finite Element Analysis , Physical Phenomena
12.
Adv Sci (Weinh) ; 11(4): e2307182, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37949680

ABSTRACT

Intracellular C-terminal cleavage of the amyloid precursor protein (APP) is elevated in the brains of Alzheimer's disease (AD) patients and produces a peptide labeled APP-C31 that is suspected to be involved in the pathology of AD. But details about the role of APP-C31 in the development of the disease are not known. Here, this work reports that APP-C31 directly interacts with the N-terminal and self-recognition regions of amyloid-ß40 (Aß40 ) to form transient adducts, which facilitates the aggregation of both metal-free and metal-bound Aß40 peptides and aggravates their toxicity. Specifically, APP-C31 increases the perinuclear and intranuclear generation of large Aß40 deposits and, consequently, damages the nucleus leading to apoptosis. The Aß40 -induced degeneration of neurites and inflammation are also intensified by APP-C31 in human neurons and murine brains. This study demonstrates a new function of APP-C31 as an intracellular promoter of Aß40 amyloidogenesis in both metal-free and metal-present environments, and may offer an interesting alternative target for developing treatments for AD that have not been considered thus far.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Humans , Mice , Animals , Amyloid beta-Protein Precursor/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Apoptosis , Promoter Regions, Genetic/genetics , Metals/toxicity
13.
Mol Cells ; 46(1): 33-40, 2023 Jan 31.
Article in English | MEDLINE | ID: mdl-36697235

ABSTRACT

RNAs are versatile molecules that are primarily involved in gene regulation and can thus be widely used to advance the fields of therapeutics and diagnostics. In particular, circular RNAs which are highly stable, have emerged as strong candidates for use on next-generation therapeutic platforms. Endogenous circular RNAs control gene regulatory networks by interacting with other biomolecules or through translation into polypeptides. Circular RNAs exhibit cell-type specific expression patterns, which can be altered in tissues and body fluids depending on pathophysiological conditions. Circular RNAs that are aberrantly expressed in diseases can function as biomarkers or therapeutic targets. Moreover, exogenous circular RNAs synthesized in vitro can be introduced into cells as therapeutic molecules to modulate gene expression networks in vivo. Depending on the purpose, synthetic circular RNA sequences can either be identical to endogenous circular RNA sequences or artificially designed. In this review, we introduce the life cycle and known functions of intracellular circular RNAs. The current stage of endogenous circular RNAs as biomarkers and therapeutic targets is also described. Finally, approaches and considerations that are important for applying the available knowledge on endogenous circular RNAs to design exogenous circular RNAs for therapeutic purposes are presented.


Subject(s)
RNA, Circular , RNA , RNA, Circular/genetics , RNA/genetics , RNA/therapeutic use , RNA/metabolism , Biomarkers , Gene Expression Regulation , Gene Regulatory Networks
14.
Exp Mol Med ; 55(7): 1283-1292, 2023 07.
Article in English | MEDLINE | ID: mdl-37430086

ABSTRACT

RNA interference mediated by small interfering RNAs (siRNAs) has been exploited for the development of therapeutics. siRNAs can be a powerful therapeutic tool because the working mechanisms of siRNAs are straightforward. siRNAs determine targets based on their sequence and specifically regulate the gene expression of the target gene. However, efficient delivery of siRNAs to the target organ has long been an issue that needs to be solved. Tremendous efforts regarding siRNA delivery have led to significant progress in siRNA drug development, and from 2018 to 2022, a total of five siRNA drugs were approved for the treatment of patients. Although all FDA-approved siRNA drugs target the hepatocytes of the liver, siRNA-based drugs targeting different organs are in clinical trials. In this review, we introduce siRNA drugs in the market and siRNA drug candidates in clinical trials that target cells in multiple organs. The liver, eye, and skin are the preferred organs targeted by siRNAs. Three or more siRNA drug candidates are in phase 2 or 3 clinical trials to suppress gene expression in these preferred organs. On the other hand, the lungs, kidneys, and brain are challenging organs with relatively few clinical trials. We discuss the characteristics of each organ related to the advantages and disadvantages of siRNA drug targeting and strategies to overcome the barriers in delivering siRNAs based on organ-specific siRNA drugs that have progressed to clinical trials.


Subject(s)
Liver , Lung , Humans , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Pharmaceutical Preparations/metabolism , RNA Interference , Lung/metabolism , Liver/metabolism
15.
Life (Basel) ; 12(9)2022 Sep 13.
Article in English | MEDLINE | ID: mdl-36143463

ABSTRACT

Long interspersed nuclear element 1 (LINE1, L1) is a retrotransposon comprising ~17% of the human genome. A subset of L1s maintains the potential to mobilize and alter the genomic landscape, consequently contributing to the change in genome integrity and gene expression. L1 retrotransposition occurs in the human brain regardless of disease status. However, in the brain of patients with various brain diseases, the expression level and copy number of L1 are significantly increased. In this review, we briefly introduce the methodologies applied to measure L1 mobility and identify genomic loci where new insertion of L1 occurs in the brain. Then, we present a list of genes disrupted by L1 transposition in the genome of patients with brain disorders. Finally, we discuss the association between genes disrupted by L1 and relative brain disorders.

16.
Nat Commun ; 12(1): 2695, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33976205

ABSTRACT

mTOR signaling, involving mTORC1 and mTORC2 complexes, critically regulates neural development and is implicated in various brain disorders. However, we do not fully understand all of the upstream signaling components that can regulate mTOR signaling, especially in neurons. Here, we show a direct, regulated inhibition of mTOR by Tanc2, an adaptor/scaffolding protein with strong neurodevelopmental and psychiatric implications. While Tanc2-null mice show embryonic lethality, Tanc2-haploinsufficient mice survive but display mTORC1/2 hyperactivity accompanying synaptic and behavioral deficits reversed by mTOR-inhibiting rapamycin. Tanc2 interacts with and inhibits mTOR, which is suppressed by mTOR-activating serum or ketamine, a fast-acting antidepressant. Tanc2 and Deptor, also known to inhibit mTORC1/2 minimally affecting neurodevelopment, distinctly inhibit mTOR in early- and late-stage neurons. Lastly, Tanc2 inhibits mTORC1/2 in human neural progenitor cells and neurons. In summary, our findings show that Tanc2 is a mTORC1/2 inhibitor affecting neurodevelopment.


Subject(s)
Brain/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Neurons/metabolism , Proteins/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Brain/embryology , Brain/growth & development , Cells, Cultured , HEK293 Cells , Humans , Immunosuppressive Agents/pharmacology , Learning Disabilities/genetics , Learning Disabilities/physiopathology , Maze Learning/drug effects , Maze Learning/physiology , Memory Disorders/genetics , Memory Disorders/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Proteins/genetics , Signal Transduction/drug effects , Sirolimus/pharmacology
17.
EMBO Mol Med ; 13(2): e12632, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33428810

ABSTRACT

Glycine transporters (GlyT1 and GlyT2) that regulate levels of brain glycine, an inhibitory neurotransmitter with co-agonist activity for NMDA receptors (NMDARs), have been considered to be important targets for the treatment of brain disorders with suppressed NMDAR function such as schizophrenia. However, it remains unclear whether other amino acid transporters expressed in the brain can also regulate brain glycine levels and NMDAR function. Here, we report that SLC6A20A, an amino acid transporter known to transport proline based on in vitro data but is understudied in the brain, regulates proline and glycine levels and NMDAR function in the mouse brain. SLC6A20A transcript and protein levels were abnormally increased in mice carrying a mutant PTEN protein lacking the C terminus through enhanced ß-catenin binding to the Slc6a20a gene. These mice displayed reduced extracellular levels of brain proline and glycine and decreased NMDAR currents. Elevating glycine levels back to normal ranges by antisense oligonucleotide-induced SLC6A20 knockdown, or the competitive GlyT1 antagonist sarcosine, normalized NMDAR currents and repetitive climbing behavior observed in these mice. Conversely, mice lacking SLC6A20A displayed increased extracellular glycine levels and NMDAR currents. Lastly, both mouse and human SLC6A20 proteins mediated proline and glycine transports, and SLC6A20 proteins could be detected in human neurons. These results suggest that SLC6A20 regulates proline and glycine homeostasis in the brain and that SLC6A20 inhibition has therapeutic potential for brain disorders involving NMDAR hypofunction.


Subject(s)
Glycine , Receptors, N-Methyl-D-Aspartate , Animals , Brain/metabolism , Glycine Plasma Membrane Transport Proteins/genetics , Glycine Plasma Membrane Transport Proteins/metabolism , Homeostasis , Membrane Transport Proteins , Mice , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
18.
Nature ; 425(6956): 415-9, 2003 Sep 25.
Article in English | MEDLINE | ID: mdl-14508493

ABSTRACT

Hundreds of small RNAs of approximately 22 nucleotides, collectively named microRNAs (miRNAs), have been discovered recently in animals and plants. Although their functions are being unravelled, their mechanism of biogenesis remains poorly understood. miRNAs are transcribed as long primary transcripts (pri-miRNAs) whose maturation occurs through sequential processing events: the nuclear processing of the pri-miRNAs into stem-loop precursors of approximately 70 nucleotides (pre-miRNAs), and the cytoplasmic processing of pre-miRNAs into mature miRNAs. Dicer, a member of the RNase III superfamily of bidentate nucleases, mediates the latter step, whereas the processing enzyme for the former step is unknown. Here we identify another RNase III, human Drosha, as the core nuclease that executes the initiation step of miRNA processing in the nucleus. Immunopurified Drosha cleaved pri-miRNA to release pre-miRNA in vitro. Furthermore, RNA interference of Drosha resulted in the strong accumulation of pri-miRNA and the reduction of pre-miRNA and mature miRNA in vivo. Thus, the two RNase III proteins, Drosha and Dicer, may collaborate in the stepwise processing of miRNAs, and have key roles in miRNA-mediated gene regulation in processes such as development and differentiation.


Subject(s)
Cell Nucleus/enzymology , Endoribonucleases/metabolism , MicroRNAs/metabolism , RNA Processing, Post-Transcriptional , Base Sequence , Cell Line , Cell Nucleus/genetics , Endoribonucleases/genetics , HeLa Cells , Humans , MicroRNAs/genetics , RNA Interference , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonuclease III
19.
Sci Adv ; 6(6): eaav7416, 2020 02.
Article in English | MEDLINE | ID: mdl-32083171

ABSTRACT

Secondary drug resistance stems from dynamic clonal evolution during the development of a prior primary resistance. This collateral type of resistance is often a characteristic of cancer recurrence. Yet, mechanisms that drive this collateral resistance and their drug-specific trajectories are still poorly understood. Using resistance selection and small-scale pharmacological screens, we find that cancer cells with primary acquired resistance to the microtubule-stabilizing drug paclitaxel often develop tolerance to epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs), leading to formation of more stable resistant cell populations. We show that paclitaxel-resistant cancer cells follow distinct selection paths under EGFR-TKIs by enriching the stemness program, developing a highly glycolytic adaptive stress response, and rewiring an apoptosis control pathway. Collectively, our work demonstrates the alterations in cellular state stemming from paclitaxel failure that result in collateral resistance to EGFR-TKIs and points to new exploitable vulnerabilities during resistance evolution in the second-line treatment setting.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drug Resistance, Neoplasm , Molecular Targeted Therapy , Paclitaxel/pharmacology , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis , Cell Line, Tumor , Cellular Senescence , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Genomics/methods , Glycolysis , Humans , Induction Chemotherapy , Models, Biological , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Paclitaxel/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Treatment Failure , Treatment Outcome
20.
Nucleic Acids Res ; 34(16): 4622-9, 2006.
Article in English | MEDLINE | ID: mdl-16963499

ABSTRACT

DGCR8/Pasha is an essential cofactor for Drosha, a nuclear RNase III that cleaves the local hairpin structures embedded in long primary microRNA transcripts (pri-miRNAs) in eukaryotes. Although our knowledge of pri-miRNA processing has significantly advanced in recent years, the precise role of DGCR8 in this pathway remains unclear. In our present study, we dissect the domains in DGCR8 that contribute to the processing of pri-miRNAs and the subcellular localization of DGCR8. Drosha is stabilized through an interaction between its middle domain and the conserved C-terminal domain of DGCR8. Furthermore, DGCR8, but not Drosha, can directly and stably interact with pri-miRNAs, and the tandem dsRNA-binding domains (dsRBDs) in DGCR8 are responsible for this recognition. Moreover, the DGCR8 N-terminal region upstream of its dsRBDs is unnecessary for pri-miRNA processing but is critical for nuclear localization. Our study thus provides further insights into the mechanism of action of the Drosha-DGCR8 complex in pri-miRNA processing.


Subject(s)
MicroRNAs/metabolism , Proteins/chemistry , RNA Processing, Post-Transcriptional , Ribonuclease III/metabolism , Amino Acid Sequence , Binding Sites , Cell Line , Cell Nucleus/chemistry , Conserved Sequence , Humans , Mutation , Protein Structure, Tertiary , Proteins/genetics , Proteins/metabolism , RNA-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL