Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
2.
bioRxiv ; 2023 Sep 14.
Article in English | MEDLINE | ID: mdl-37745332

ABSTRACT

Mutations in the GBA1 gene have been identified as a prevalent genetic risk factor for Parkinson's disease (PD). GBA1 mutations impair enzymatic activity, leading to lysosomal dysfunction and elevated levels of α-synuclein (α-syn). While most research has primarily focused on GBA1's role in promoting synucleinopathy, emerging evidence suggests that neuroinflammation may be a key pathogenic alteration caused by GBA1 deficiency. To examine the molecular mechanism underlying GBA1 deficiency-mediated neuroinflammation, we generated Gba1 E326K knock-in (KI) mice using the CRISPR/Cas9 technology, which is linked to an increased risk of PD and dementia with Lewy bodies (DLB). In the ventral midbrain and hippocampus of 24-month-old Gba1 E326K KI mice, we found a moderate decline in GBA1 enzymatic activity, a buildup of glucosylceramide, and an increase in microglia density. Furthermore, we observed increased levels of pro-inflammatory cytokines and formation of reactive astrocytes in primary microglia and astrocytes, respectively, cultured from Gba1 E326K KI mice following treatment with pathologic α-syn preformed fibrils (PFF). Additionally, the gut inoculation of α-syn PFF in Gba1 E326K KI mice significantly enhanced the accumulation of Lewy bodies in the dentate gyrus of the hippocampus, accompanied by aggravated neuroinflammation and exacerbated non-motor symptoms. This research significantly enhances our understanding of the Gba1 E326K mutation's involvement in neuroinflammation and the cell-to-cell transmission of pathogenic α-syn in the brain, thereby opening new therapeutic avenues.

3.
Cells ; 11(24)2022 12 08.
Article in English | MEDLINE | ID: mdl-36552734

ABSTRACT

Non-receptor tyrosine kinase, c-Abl plays a role in the pathogenesis of several neurodegenerative disorders such as Alzheimer's disease and Parkinson's disease. Here, we found that TDP-43, which was one of the main proteins comprising pathological deposits in amyotrophic lateral sclerosis (ALS), is a novel substrate for c-Abl. The phosphorylation of tyrosine 43 of TDP-43 by c-Abl led to increased TDP-43 levels in the cytoplasm and increased the formation of G3BP1-positive stress granules in SH-SY5Y cells. The kinase-dead mutant of c-Abl had no effect on the cytoplasmic localization of TDP-43. The expression of phosphor-mimetic mutant Y43E of TDP-43 in primary cortical neurons accumulated the neurite granule. Furthermore, the phosphorylation of TDP-43 at tyrosine 43 by c-Abl promoted the aggregation of TDP-43 and increased neuronal cell death in primary cortical neurons, but not in c-Abl-deficient primary cortical neurons. Identification of c-Abl as the kinase of TDP43 provides new insight into the pathogenesis of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Proto-Oncogene Proteins c-abl , Humans , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Neuroblastoma , Phosphorylation , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Tyrosine/metabolism
4.
Oxid Med Cell Longev ; 2022: 4392256, 2022.
Article in English | MEDLINE | ID: mdl-35979399

ABSTRACT

The regulation of collagen synthesis, which occurs in fibroblasts in the dermal layer, is a key process in dermis regeneration and skin reconstruction. Herein, we investigated whether Aronia melanocarpa extract affects the human skin condition. We focused on type I collagen synthesis using two different types of model systems: a monolayer of cells and a bioprinted 3D dermal equivalent. The Aronia extract showed no cytotoxicity and increased cell proliferation in neonatal human dermal fibroblasts. Treatment with Aronia extract increased the transcription of COL1A1 mRNA in direct proportion to the extract concentration without causing a decrease in COL1A1 mRNA degradation. Additionally, the Aronia extract inhibited the expression of MMP1 and MMP3, and an increase in type I collagen was observed along with a decrease in MMP1 protein. We also fabricated dermal equivalents from type I collagen (the major component of the dermis) and dermal fibroblasts by bioprinting. In the 3D dermis model, the compressive modulus directly affected by collagen synthesis increased in direct proportion to the Aronia extract concentration, and expression levels of MMP1 and MMP3 decreased in exactly inverse proportion to its concentration. The findings that the Aronia extract increases synthesis of type I collagen and decreases MMP1 and MMP3 expression suggest that this extract may be useful for the treatment of damaged or aged skin.


Subject(s)
Matrix Metalloproteinase 1 , Photinia , Aged , Cells, Cultured , Collagen Type I/metabolism , Fibroblasts/metabolism , Humans , Infant, Newborn , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/metabolism , Photinia/metabolism , Skin/metabolism
5.
Int J Mol Sci ; 22(11)2021 May 23.
Article in English | MEDLINE | ID: mdl-34071140

ABSTRACT

THeterogeneous nuclear ribonucleoprotein (HNRNP) A1 is the most abundant and ubiquitously expressed member of the HNRNP protein family. In recent years, it has become more evident that HNRNP A1 contributes to the development of neurodegenerative diseases. However, little is known about the underlying role of HNRNP A1 in cancer development. Here, we report that HNRNP A1 expression is significantly increased in lung cancer tissues and is negatively correlated with the overall survival of patients with lung cancer. Additionally, HNRNP A1 positively regulates vaccinia-related kinase 1 (VRK1) translation via binding directly to the 3' untranslated region (UTR) of VRK1 mRNA, thus increasing cyclin D1 (CCND1) expression by VRK1-mediated phosphorylation of the cAMP response element-binding protein (CREB). Furthermore, HNRNP A1 binding to the cis-acting region of the 3'UTR of VRK1 mRNA contributes to increased lung cancer cell proliferation. Thus, our study unveils a novel role of HNRNP A1 in lung carcinogenesis via post-transcriptional regulation of VRK1 expression and suggests its potential as a therapeutic target for patients with lung cancer.


Subject(s)
Heterogeneous Nuclear Ribonucleoprotein A1/physiology , Intracellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/pathology , Neoplasm Proteins/physiology , Protein Biosynthesis , Protein Serine-Threonine Kinases/genetics , 3' Untranslated Regions , Base Sequence , CRISPR-Cas Systems , Cell Cycle , Cell Line , Cyclin D1/biosynthesis , Cyclin D1/genetics , Eukaryotic Initiation Factor-3/metabolism , Gene Expression Regulation, Neoplastic , Genes, Reporter , Heterogeneous Nuclear Ribonucleoprotein A1/chemistry , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , Lung Neoplasms/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Protein Binding , Protein Domains , Protein Interaction Mapping , Protein Serine-Threonine Kinases/biosynthesis , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Sequence Deletion , Up-Regulation
6.
Sci Adv ; 6(47)2020 11.
Article in English | MEDLINE | ID: mdl-33219033

ABSTRACT

The AMPA receptor subunit GluA1 is essential for induction of synaptic plasticity. While various regulatory mechanisms of AMPA receptor expression have been identified, the underlying mechanisms of GluA1 protein synthesis are not fully understood. In neurons, axonal and dendritic mRNAs have been reported to be translated in a cap-independent manner. However, molecular mechanisms of cap-independent translation of synaptic mRNAs remain largely unknown. Here, we show that GluA1 mRNA contains an internal ribosome entry site (IRES) in the 5'UTR. We also demonstrate that heterogeneous nuclear ribonucleoprotein (hnRNP) A2/B1 interacts with GluA1 mRNA and mediates internal initiation of GluA1 Brain-derived neurotrophic factor (BDNF) stimulation increases IRES-mediated GluA1 translation via up-regulation of HNRNP A2/B1. Moreover, BDNF-induced GluA1 expression and dendritic spine density were significantly decreased in neurons lacking hnRNP A2/B1. Together, our data demonstrate that IRES-mediated translation of GluA1 mRNA is a previously unidentified feature of local expression of the AMPA receptor.

7.
J Biol Chem ; 294(19): 7682-7691, 2019 05 10.
Article in English | MEDLINE | ID: mdl-30948510

ABSTRACT

Most living creatures have a circadian rhythm that is generated by a precisely regulated transcriptional-translational feedback loop of clock genes. Brain and muscle ARNT-like 1 (BMAL1) is one of the core clock genes and transcription factors that represents a positive arm of this autoregulatory circadian clock system. Despite the indispensable role of BMAL1 in the circadian rhythm, the molecular mechanisms underlying translational control of BMAL1 are largely unknown. Here, using murine NIH-3T3 cells, gene constructs, and a variety of biochemical approaches, including RNAi- and luciferase reporter gene-based assays, along with immunoblotting, in vitro transcription, quantitative real-time PCR, and real-time bioluminescence experiments, we show that translation of Bmal1 is negatively regulated by an RNA-binding protein, heterogeneous nuclear ribonucleoprotein Q (hnRNP Q). Interestingly, we found that hnRNP Q rhythmically binds to a specific region of the Bmal1 mRNA 5' UTR and controls its time-dependent expression. Moreover, we demonstrate that knockdown of hnRNP Q modulates BMAL1 protein oscillation amplitude without affecting mRNA rhythmic patterns. Furthermore, hnRNP Q depletion increases the mRNA oscillation amplitudes of BMAL1-regulated target genes. Together, our results suggest that hnRNP Q plays a pivotal role in both Bmal1 translation and BMAL1-regulated gene expression.


Subject(s)
5' Untranslated Regions , ARNTL Transcription Factors/biosynthesis , Gene Expression Regulation , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , ARNTL Transcription Factors/genetics , Animals , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Mice , NIH 3T3 Cells , Protein Transport/genetics , RNA, Messenger/genetics
8.
Cell Death Dis ; 10(2): 49, 2019 01 18.
Article in English | MEDLINE | ID: mdl-30718468

ABSTRACT

Proper wiring between neurons is indispensable for proper brain function. From the early developmental stage, axons grow and navigate to connect to targets according to specific guidance cues. The accuracy of axonal outgrowth and navigation are controlled by a variety of genes, and mutations and/or deficiencies in these genes are closely related to several brain disorders, such as autism. DSCR1 is one of these genes and regulates actin filament formation in axons. Thus, identifying the detailed regulatory mechanisms of DSCR1 expression is crucial for the understanding of the axon development of neurons; however, these regulatory mechanisms of DSCR1 remain unknown. Here, we discovered that mRNA encoding the DSCR1 isoform DSCR1.4 is present and mainly translated by the cap-independent initiation mechanisms in both the soma and axons of hippocampal neurons. We found that translation of DSCR1.4 mRNA is enhanced by death-associated protein 5 (DAP5), which can bind to DSCR1.4 5'UTR. BDNF-stimulus induced an increase in DAP5 expression and the cap-independent translation efficiency of DSCR1.4 mRNA in axon as well as soma. Furthermore, we showed the importance of the cap-independent translation of DSCR1.4 on enhancement of DSCR1.4 expression by BDNF-stimulus and axonal outgrowth of hippocampal neurons. Our findings suggest a new translational regulatory mechanism for DSCR1.4 expressions and a novel function of DAP5 as a positive regulator of DSCR1.4 mRNA translation induced in soma and axon of hippocampal neurons.


Subject(s)
Axons/metabolism , DNA-Binding Proteins/genetics , Eukaryotic Initiation Factor-4G/metabolism , Hippocampus/metabolism , Muscle Proteins/genetics , Neurons/metabolism , RNA, Messenger/metabolism , 5' Untranslated Regions , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Eukaryotic Initiation Factor-4G/genetics , Hippocampus/cytology , Humans , Mice , Muscle Proteins/metabolism , Neurons/cytology , Protein Isoforms , RNA, Messenger/genetics , Transfection
9.
J Neurochem ; 149(3): 413-426, 2019 05.
Article in English | MEDLINE | ID: mdl-30488434

ABSTRACT

Misfolded proteins with abnormal polyglutamine (polyQ) expansion cause neurodegenerative disorders, including Huntington's disease. Recently, it was found that polyQ aggregates accumulate as a result of vaccinia-related kinase 2 (VRK2)-mediated degradation of TCP-1 ring complex (TRiC)/chaperonin-containing TCP-1 (CCT), which has an essential role in the prevention of polyQ protein aggregation and cytotoxicity. The levels of VRK2 are known to be much higher in actively proliferating cells but are maintained at a low level in the brain via an unknown mechanism. Here, we found that basal levels of neuronal cell-specific VRK2 mRNA are maintained by post-transcriptional, rather than transcriptional, regulation. Moreover, heterogeneous nuclear ribonucleoprotein Q (HNRNP Q) specifically binds to the 3'untranslated region of VRK2 mRNA in neuronal cells to reduce the mRNA stability. As a result, we found a dramatic decrease in CCT4 protein levels in response to a reduction in HNRNP Q levels, which was followed by an increase in polyQ aggregation in human neuroblastoma cells and mouse cortical neurons. Taken together, these results provide new insights into how neuronal HNRNP Q decreases VRK2 mRNA stability and contributes to the prevention of Huntington's disease, while also identifying new prognostic markers of HD.


Subject(s)
Gene Expression Regulation/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Protein Serine-Threonine Kinases/biosynthesis , Animals , Brain/metabolism , Mice , Mice, Inbred C57BL , Peptides/metabolism , RNA Processing, Post-Transcriptional
10.
J Cell Sci ; 132(2)2019 01 18.
Article in English | MEDLINE | ID: mdl-30584064

ABSTRACT

A large number of neuronal proteins must show correct spatiotemporal localization in order to carry out their critical functions. The mRNA transcript for the somatodendritic protein activity-regulated cytoskeleton-associated protein (Arc; also known as Arg3.1) contains two conserved introns in the 3' untranslated region (UTR), and was proposed to be a natural target for nonsense-mediated mRNA decay (NMD). However, a well-known NMD component Upf1 has differential roles in transcriptional and translational regulation of Arc gene expression. Specifically, Upf1 suppresses Arc transcription by enhancing destabilization of mRNAs encoding various transcription factors, including Mef2a. Upf1 also binds to the Arc 3'UTR, resulting in suppression of translation. Surprisingly, the Arc transcript escapes from Upf1-mediated NMD by binding to Ago2 (also known as miRISC), which blocks NMD and further suppresses Arc mRNA translation. Upf1 knockdown triggered sustained Arc expression, which contributes to Cofilin (also known as Cfl1) hyperphosphorylation and abnormal neuronal outgrowth and branching. Collectively, these data reveal that multiple levels of Upf1-mediated inhibition of Arc gene expression may allow neurons to more effectively respond to changes in neuronal activity.


Subject(s)
Cytoskeletal Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurites/metabolism , Nonsense Mediated mRNA Decay , Trans-Activators/metabolism , Transcription, Genetic , Animals , Cell Line , Cofilin 1/genetics , Cofilin 1/metabolism , Cytoskeletal Proteins/genetics , Mice , Nerve Tissue Proteins/genetics , Trans-Activators/genetics
11.
Diabetes Obes Metab ; 20(7): 1688-1701, 2018 07.
Article in English | MEDLINE | ID: mdl-29516607

ABSTRACT

AIMS: Although peroxisome proliferator-activated receptors (PPARs)α/γ dual agonists can be beneficial for treatment of dyslipidemia in patients with type 2 diabetes, their use is limited owing to various side effects, including body weight gain, edema, and heart failure. We aimed to demonstrate that amodiaquine, an antimalarial agent, has potential as a PPARα/γ dual agonist with low risk of adverse effects. METHODS: We screened a Prestwick library (Prestwick Chemical; Illkirch, France) to identify novel PPARα/γ dual agonists and selected amodiaquine (4-[(7-chloroquinolin-4-yl)amino]-2-[(diethylamino)methyl]phenol), which activated both PPAR-α & -γ, for further investigation. We performed both in vitro, including glucose uptake assay and fatty acid oxidation assay, and in vivo studies to elucidate the anti-diabetic and anti-obesity effects of amodiaquine. RESULTS: Amodiaquine selectively activated the transcriptional activities of PPARα/γ and enhanced both fatty acid oxidation and glucose uptake without altering insulin secretion in vitro. In high-fat diet-induced obese and genetically modified obese/diabetic mice, amodiaquine not only remarkably ameliorated insulin resistance, hyperlipidemia, and fatty liver but also decreased body weight gain. CONCLUSION: Our findings suggest that amodiaquine exerts beneficial effects on glucose and lipid metabolism by concurrent activation of PPARα/γ. Furthermore, amodiaquine acts as an alternative insulin-sensitizing agent with a positive influence on lipid metabolism and has potential to prevent and treat type 2 diabetes while reducing the risk of lipid abnormalities.


Subject(s)
Amodiaquine/pharmacology , Antimalarials/pharmacology , Blood Glucose/drug effects , Insulin Resistance , Lipid Metabolism/drug effects , Liver/drug effects , PPAR alpha/agonists , PPAR gamma/agonists , 3T3-L1 Cells , Animals , Blood Glucose/metabolism , Body Weight , Cell Proliferation , Diet, High-Fat , Disease Models, Animal , Fatty Acids/metabolism , Fatty Liver , Hyperlipidemias , In Vitro Techniques , Liver/metabolism , Mice , Mice, Obese , Oxidation-Reduction , Triglycerides/metabolism
12.
Oncotarget ; 8(31): 51108-51122, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28881634

ABSTRACT

The tumor suppressor p53 is an essential gene in the induction of cell cycle arrest, DNA repair, and apoptosis. p53 protein is induced under cellular stress, blocking cell cycle progression and inducing DNA repair. Under DNA damage conditions, it has been reported that post-transcriptional regulation of p53 mRNA contributes to the increase in p53 protein level. Here we demonstrate that heterogeneous nuclear ribonucleoprotein (hnRNP) L enhances p53 mRNA translation. We found that hnRNP L is increased and binds to the 5'UTR of p53 mRNA in response to DNA damage. Increased hnRNP L caused enhancement of p53 mRNA translation. Conversely, p53 protein levels were decreased following hnRNP L knock-down, rendering them resistant to apoptosis and arrest in the G2/M phase after DNA damage. Thus, our findings suggest that hnRNP L functions as a positive regulator of p53 translation and promotes cell cycle arrest and apoptosis.

13.
Mol Cells ; 40(9): 621-631, 2017 Sep 30.
Article in English | MEDLINE | ID: mdl-28927264

ABSTRACT

Vaccinia-related kinase 1 (VRK1) and VRK3 are members of the VRK family of serine/threonine kinases and are principally localized in the nucleus. Despite the crucial roles of VRK1/VRK3 in physiology and disease, the molecular and functional interactions of VRK1/VRK3 are poorly understood. Here, we identified over 200 unreported VRK1/VRK3-interacting candidate proteins by affinity purification and LC-MS/MS. The networks of VRK1 and VRK3 interactomes were found to be associated with important biological processes such as the cell cycle, DNA repair, chromatin assembly, and RNA processing. Interactions of interacting proteins with VRK1/VRK3 were confirmed by biochemical assays. We also found that phosphorylations of XRCC5 were regulated by both VRK1/VRK3, and that of CCNB1 was regulated by VRK3. In liver cancer cells and tissues, VRK1/VRK3 were highly upregulated and its depletion affected cell cycle progression in the different phases. VRK3 seemed to affect S phase progression and G2 or M phase entry and exit, whereas VRK1 affects G1/S transition in the liver cancer, which could be explained by different interacting candidate proteins. Thus, this study not only provides a resource for investigating the unidentified functions of VRK1/VRK3, but also an insight into the regulatory roles of VRK1/VRK3 in biological processes.


Subject(s)
Cell Proliferation/genetics , Intracellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cyclin B1/genetics , Gene Expression Regulation/genetics , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Ku Autoantigen/genetics , Liver Neoplasms/pathology , Phosphorylation , Protein Interaction Maps , Protein Serine-Threonine Kinases/metabolism
14.
J Exp Med ; 214(10): 2947-2966, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28899869

ABSTRACT

Vaccinia-related kinases (VRKs) are multifaceted serine/threonine kinases that play essential roles in various aspects of cell signaling, cell cycle progression, apoptosis, and neuronal development and differentiation. However, the neuronal function of VRK3 is still unknown despite its etiological potential in human autism spectrum disorder (ASD). Here, we report that VRK3-deficient mice exhibit typical symptoms of autism-like behavior, including hyperactivity, stereotyped behaviors, reduced social interaction, and impaired context-dependent spatial memory. A significant decrease in dendritic spine number and arborization were identified in the hippocampus CA1 of VRK3-deficient mice. These mice also exhibited a reduced rectification of AMPA receptor-mediated current and changes in expression of synaptic and signaling proteins, including tyrosine receptor kinase B (TrkB), Arc, and CaMKIIα. Notably, TrkB stimulation with 7,8-dihydroxyflavone reversed the altered synaptic structure and function and successfully restored autism-like behavior in VRK3-deficient mice. These results reveal that VRK3 plays a critical role in neurodevelopmental disorders and suggest a potential therapeutic strategy for ASD.


Subject(s)
Autistic Disorder/etiology , Protein Serine-Threonine Kinases/deficiency , Receptor, trkB/physiology , Animals , CA1 Region, Hippocampal/pathology , Female , Flavanones/pharmacology , Hyperkinesis/etiology , Male , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , Receptor, trkB/drug effects , Receptor, trkB/metabolism , Social Behavior , Stereotyped Behavior
15.
Sci Rep ; 7(1): 6237, 2017 07 24.
Article in English | MEDLINE | ID: mdl-28740165

ABSTRACT

Robust mitochondrial respiration provides energy to support physical performance and physiological well-being, whereas mitochondrial malfunction is associated with various pathologies and reduced longevity. In the current study, we tested whether myricetin, a natural flavonol with diverse biological activities, may impact mitochondrial function and longevity. The mice were orally administered myricetin (50 mg/kg/day) for 3 weeks. Myricetin significantly potentiated aerobic capacity in mice, as evidenced by their increased running time and distance. The elevated mitochondrial function was associated with induction of genes for oxidative phosphorylation and mitochondrial biogenesis in metabolically active tissues. Importantly, myricetin treatment led to decreased PGC-1α acetylation through SIRT1 activation. Furthermore, myricetin significantly improved the healthspan and lifespan of wild-type, but not Sir-2.1-deficient, C. elegans. These results demonstrate that myricetin enhances mitochondrial activity, possibly by activating PGC-1α and SIRT1, to improve physical endurance, strongly suggesting myricetin as a mitochondria-activating agent.


Subject(s)
Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Longevity , Mitochondria/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Physical Endurance/drug effects , Sirtuin 1/metabolism , Animals , Caenorhabditis elegans , Male , Mice , Mice, Inbred ICR , Mitochondria/drug effects , Organelle Biogenesis , Oxidative Phosphorylation , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Sirtuin 1/genetics
16.
Sci Rep ; 7: 42882, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28220845

ABSTRACT

Nuclear factor, interleukin 3, regulated (Nfil3, also known as E4 Promoter-Binding Protein 4 (E4BP4)) protein is a transcription factor that binds to DNA and generally represses target gene expression. In the circadian clock system, Nfil3 binds to a D-box element residing in the promoter of clock genes and contributes to their robust oscillation. Here, we show that the 5'-untranslated region (5'-UTR) of Nfil3 mRNA contains an internal ribosome entry site (IRES) and that IRES-mediated translation occurs in a phase-dependent manner. We demonstrate that heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1) binds to a specific region of Nfil3 mRNA and regulates IRES-mediated translation. Knockdown of hnRNP A1 almost completely abolishes protein oscillation without affecting mRNA oscillation. Moreover, we observe that intracellular calcium levels, which are closely related to bone formation, depend on Nfil3 levels in osteoblast cell lines. We suggest that the 5'-UTR mediated cap-independent translation of Nfil3 mRNA contributes to the rhythmic expression of Nfil3 by interacting with the RNA binding protein hnRNP A1. These data provide new evidence that the posttranscriptional regulation of clock gene expression is important during bone metabolism.


Subject(s)
Basic-Leucine Zipper Transcription Factors/biosynthesis , Heterogeneous Nuclear Ribonucleoprotein A1/metabolism , 5' Untranslated Regions , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Calcium/metabolism , Cell Line , Circadian Clocks , Heterogeneous Nuclear Ribonucleoprotein A1/antagonists & inhibitors , Heterogeneous Nuclear Ribonucleoprotein A1/genetics , Internal Ribosome Entry Sites , Mice , Osteogenesis , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Regulatory Elements, Transcriptional , Ribosomes/chemistry , Ribosomes/metabolism
17.
PLoS One ; 11(11): e0165835, 2016.
Article in English | MEDLINE | ID: mdl-27824900

ABSTRACT

The role of Sirtuin 6 (SIRT6) as a tumor suppressor or oncogene in liver cancer remains controversial. Thus, we identified the specific role of SIRT6 in the progression of hepatocellular carcinoma (HCC). SIRT6 expression was significantly higher in HCC cell lines and HCC tissues from 138 patients than in an immortalized hepatocyte cell line, THLE-2 and non-tumor tissues, respectively. SIRT6 knockdown by shRNA suppressed the growth of HCC cells and inhibited HCC tumor growth in vivo. In addition, SIRT6 silencing significantly prevented the growth of HCC cell lines by inducing cellular senescence in the p16/Rb- and p53/p21-pathway independent manners. Microarray analysis revealed that the expression of genes involved in nucleosome assembly was apparently altered in SIRT6-depleted Hep3B cells. SIRT6 knockdown promoted G2/M phase arrest and downregulation of genes encoding histone variants associated with nucleosome assembly, which could be attributed to DNA damage. Taken together, our findings suggest that SIRT6 acts as a tumor promoter by preventing DNA damage and cellular senescence, indicating that SIRT6 represents a potential therapeutic target for the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Cellular Senescence/physiology , DNA Damage/physiology , Liver Neoplasms/physiopathology , Sirtuins/physiology , Animals , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/physiology , Gene Knockdown Techniques , Humans , Liver Neoplasms/metabolism , Male , Mice , Mice, Nude , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Sirtuins/deficiency
18.
Sci Rep ; 6: 29097, 2016 07 05.
Article in English | MEDLINE | ID: mdl-27377031

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder caused by an abnormal expansion of polyglutamine repeats in the N-terminal of huntingtin. The amount of aggregate-prone protein is controlled by various mechanisms, including molecular chaperones. Vaccinia-related kinase 2 (VRK2) is known to negatively regulate chaperonin TRiC, and VRK2-facilitated degradation of TRiC increases polyQ protein aggregation, which is involved in HD. We found that VRK2 activity was negatively controlled by glycogen synthase kinase 3ß (GSK3ß). GSK3ß directly bound to VRK2 and inhibited the catalytic activity of VRK2 in a kinase activity-independent manner. Furthermore, GSK3ß increased the stability of TRiC and decreased the formation of HttQ103-GFP aggregates by inhibiting VRK2. These results indicate that GSK3ß signaling may be a regulatory mechanism of HD progression and suggest targets for further therapeutic trials for HD.


Subject(s)
Chaperonin Containing TCP-1/genetics , Glycogen Synthase Kinase 3 beta/genetics , Huntingtin Protein/genetics , Huntington Disease/genetics , Protein Serine-Threonine Kinases/genetics , Glycogen Synthase Kinase 3 beta/chemistry , Humans , Huntingtin Protein/chemistry , Huntington Disease/metabolism , Huntington Disease/pathology , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Phosphorylation , Protein Aggregates/genetics , Protein Binding , Protein Serine-Threonine Kinases/chemistry
19.
Adv Mater ; 28(21): 3978, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27246918

ABSTRACT

Quantitative probing of the Cu(2+) ions naturally present in single living cells is accomplished by a probe made from a quantum-dot-embedded-nanowire waveguide. After inserting the active nanowire-based waveguide probe into single living cells, J. H. Je and co-workers directly observe photoluminescence (PL) quenching of the embedded quantum dots by the Cu(2+) ions diffused into the probe as described on page 4071. This results in quantitative measurement of intracellular Cu(2+) ions.


Subject(s)
Copper/chemistry , Nanowires/chemistry , Cations, Divalent , Cell Survival , Quantum Dots
20.
Adv Mater ; 28(21): 4071-6, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27027298

ABSTRACT

Quantitative probing of Cu(2+) ions naturally present in single living cells is realized by developing a quantum-dot-embedded nanowire-waveguide probe. The intracellular Cu(2+) ion concentration is quantified by direct monitoring of photoluminescence quenching during the insertion of the nanowire in a living neuron. The measured intracellular Cu(2+) ion concentration is 3.34 ± 1.04 × 10(-6) m (mean ± s.e.m.) in single hippocampal neurons.


Subject(s)
Copper/chemistry , Cations, Divalent , Nanowires , Quantum Dots
SELECTION OF CITATIONS
SEARCH DETAIL
...