Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 165
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Pharmacol ; 104(5): 214-229, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37595967

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is a severe liver metabolic disorder, however, there are still no effective and safe drugs for its treatment. Previous clinical trials used various therapeutic approaches to target individual pathologic mechanisms, but these approaches were unsuccessful because of the complex pathologic causes of NASH. Combinatory therapy in which two or more drugs are administered simultaneously to patients with NASH, however, carries the risk of side effects associated with each individual drug. To solve this problem, we identified gossypetin as an effective dual-targeting agent that activates AMP-activated protein kinase (AMPK) and decreases oxidative stress. Administration of gossypetin decreased hepatic steatosis, lobular inflammation and liver fibrosis in the liver tissue of mice with choline-deficient high-fat diet and methionine-choline deficient diet (MCD) diet-induced NASH. Gossypetin functioned directly as an antioxidant agent, decreasing hydrogen peroxide and palmitate-induced oxidative stress in the AML12 cells and liver tissue of MCD diet-fed mice without regulating the antioxidant response factors. In addition, gossypetin acted as a novel AMPK activator by binding to the allosteric drug and metabolite site, which stabilizes the activated structure of AMPK. Our findings demonstrate that gossypetin has the potential to serve as a novel therapeutic agent for nonalcoholic fatty liver disease /NASH. SIGNIFICANCE STATEMENT: This study demonstrates that gossypetin has preventive effect to progression of nonalcoholic steatohepatitis (NASH) as a novel AMP-activated protein kinase (AMPK) activator and antioxidants. Our findings indicate that simultaneous activation of AMPK and oxidative stress using gossypetin has the potential to serve as a novel therapeutic approach for nonalcoholic fatty liver disease /NASH patients.


Subject(s)
Non-alcoholic Fatty Liver Disease , Humans , Animals , Mice , Non-alcoholic Fatty Liver Disease/drug therapy , AMP-Activated Protein Kinases/metabolism , Antioxidants/metabolism , Liver/metabolism , Oxidative Stress , Choline/metabolism , Choline/pharmacology , Choline/therapeutic use , Methionine/metabolism , Methionine/pharmacology , Mice, Inbred C57BL , Disease Models, Animal
2.
EMBO J ; 37(5)2018 03 01.
Article in English | MEDLINE | ID: mdl-29440227

ABSTRACT

In neuronal development, dynamic rearrangement of actin promotes axonal growth cone extension, and spatiotemporal translation of local mRNAs in response to guidance cues directs axonal growth cone steering, where cofilin plays a critical role. While regulation of cofilin activity is well studied, regulatory mechanism for cofilin mRNA translation in neurons is unknown. In eukaryotic cells, proteins can be synthesized by cap-dependent or cap-independent mechanism via internal ribosome entry site (IRES)-mediated translation. IRES-mediated translation has been reported in various pathophysiological conditions, but its role in normal physiological environment is poorly understood. Here, we report that 5'UTR of cofilin mRNA contains an IRES element, and cofilin is predominantly translated by IRES-mediated mechanism in neurons. Furthermore, we show that IRES-mediated translation of cofilin is required for both axon extension and axonal growth cone steering. Our results provide new insights into the function of IRES-mediated translation in neuronal development.


Subject(s)
Axons/physiology , Cofilin 1/genetics , Growth Cones/physiology , Internal Ribosome Entry Sites/genetics , Neurogenesis/genetics , 5' Untranslated Regions/genetics , Animals , Brain/embryology , CRISPR-Cas Systems , Cell Line , Cell Proliferation/genetics , Cofilin 1/metabolism , Mice , Protein Biosynthesis/genetics , RNA, Messenger/genetics
3.
J Cell Sci ; 133(21)2020 11 09.
Article in English | MEDLINE | ID: mdl-33067254

ABSTRACT

Vaccinia-related kinase 3 (VRK3) has been reported to be a negative regulator of ERK (ERK1 and ERK2; also known as MAPK3 and MAPK1, respectively) that protects cells from persistent ERK activation and inhibits ERK-dependent apoptosis. Here we report that the E3 ubiquitin-protein ligase RNF144a promotes the degradation of VRK3 via polyubiquitylation and thus affects VRK3-mediated ERK activity. Under oxidative stress, VRK3 migrates from the nucleus to the cytoplasm, which increases its chance of interacting with RNF144a, thereby promoting the degradation of VRK3. Overexpression of RNF144a increases ERK activity via downregulation of VRK3 and promotes ERK-dependent apoptosis. In contrast, depletion of RNF144a increases the protein level of VRK3 and protects cells from excessive ERK activity. These findings suggest that VRK3 protects cells by suppressing oxidative stress-induced ERK, and that RNF144a sensitively regulates this process.


Subject(s)
Vaccinia , Apoptosis/genetics , Carrier Proteins/metabolism , Down-Regulation/genetics , Humans , Oxidative Stress/genetics , Phosphorylation , Protein Serine-Threonine Kinases , Ubiquitin-Protein Ligases/genetics
4.
FASEB J ; 35(4): e21507, 2021 04.
Article in English | MEDLINE | ID: mdl-33724572

ABSTRACT

Retinoic acid-related orphan receptor γ (RORγ) maintains the circadian rhythms of its downstream genes. However, the mechanism behind the transcriptional activation of RORγ itself remains unclear. Here, we demonstrate that transcription of RORγ is activated by heterogeneous nuclear ribonucleoprotein K (hnRNP K) via the poly(C) motif within its proximal promoter. Interestingly, we confirmed the binding of endogenous hnRNP K within RORγ1 and RORγ2 promoter along with the recruitment of RNA polymerase 2 through chromatin immunoprecipitation (ChIP). Furthermore, an assay for transposase accessible chromatin (ATAC)-qPCR showed that hnRNP K induced higher chromatin accessibility within the RORγ1 and RORγ2 promoter. Then we found that the knockdown of hnRNP K lowers RORγ mRNA oscillation amplitude in both RORγ and RORγ-dependent metabolic genes. Moreover, we demonstrated that time-dependent extracellular signal-regulated kinase (ERK) activation controls mRNA oscillation of RORγ and RORγ-dependent metabolic genes through hnRNP K. Taken together, our results provide new insight into the regulation of RORγ by hnRNP K as a transcriptional activator, along with its physiological significance in metabolism.


Subject(s)
Chromatin/metabolism , Circadian Rhythm/physiology , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Animals , Chromatin Immunoprecipitation/methods , Circadian Rhythm/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Mice , Transcription Factors/metabolism , Transcriptional Activation/physiology
5.
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
6.
Int J Mol Sci ; 22(22)2021 Nov 22.
Article in English | MEDLINE | ID: mdl-34830467

ABSTRACT

Although proliferation of keratinocytes, a major type of skin cells, is a key factor in maintaining the function of skin, their ability to proliferate tends to diminish with age. To solve such a problem, researchers in medical and skin cosmetic fields have tried to utilize epidermal growth factor (EGF), but achieved limited success. Therefore, a small natural compound that can mimic the activity of EGF is highly desired in both medical and cosmetic fields. Here, using the modified biosensor system, we observed that natural small-compound isoprocurcumenol, which is a terpenoid molecule derived from turmeric, can activate EGFR signaling. It increased the phosphorylation of ERK and AKT, and upregulated the expression of genes related to cell growth and proliferation, such as c-myc, c-jun, c-fos, and egr-1. In addition, isoprocurcumenol induced the proliferation of keratinocytes in both physical and UVB-induced cellular damage, indicative of its function in skin regeneration. These findings reveal that EGF-like isoprocurcumenol promotes the proliferation of keratinocytes and further suggest its potential as an ingredient for medical and cosmetics use.


Subject(s)
Cell Proliferation/drug effects , Regeneration/drug effects , Sesquiterpenes/pharmacology , Transcriptional Activation/drug effects , Cell Line , Epidermal Growth Factor/genetics , ErbB Receptors/genetics , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Keratinocytes/metabolism , MAP Kinase Signaling System/drug effects , Phosphorylation/drug effects , Protein Binding/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-myc/genetics , Sesquiterpenes/chemistry , Signal Transduction/drug effects , Skin/growth & development , Skin/metabolism , Wound Healing/drug effects
7.
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
8.
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
9.
Int J Mol Sci ; 21(10)2020 May 18.
Article in English | MEDLINE | ID: mdl-32443632

ABSTRACT

Neuronal axons are guided to their target during the development of the brain. Axon guidance allows the formation of intricate neural circuits that control the function of the brain, and thus the behavior. As the axons travel in the brain to find their target, they encounter various axon guidance cues, which interact with the receptors on the tip of the growth cone to permit growth along different signaling pathways. Although many scientists have performed numerous studies on axon guidance signaling pathways, we still have an incomplete understanding of the axon guidance system. Lately, studies on axon guidance have shifted from studying the signal transduction pathways to studying other molecular features of axon guidance, such as the gene expression. These new studies present evidence for different molecular features that broaden our understanding of axon guidance. Hence, in this review we will introduce recent studies that illustrate different molecular features of axon guidance. In particular, we will review literature that demonstrates how axon guidance cues and receptors regulate local translation of axonal genes and how the expression of guidance cues and receptors are regulated both transcriptionally and post-transcriptionally. Moreover, we will highlight the pathological relevance of axon guidance molecules to specific diseases.


Subject(s)
Axon Guidance , Axons/metabolism , Protein Biosynthesis , RNA-Binding Proteins/metabolism , Animals , Gene Expression Regulation , Humans , Signal Transduction
10.
Int J Mol Sci ; 21(18)2020 Sep 21.
Article in English | MEDLINE | ID: mdl-32967200

ABSTRACT

Circadian oscillations of mRNAs and proteins are the main features of circadian clock genes. Among them, Period1 (Per1) is a key component in negative-feedback regulation, which shows a robust diurnal oscillation and the importance of circadian rhythm and translational regulation of circadian clock genes has been recognized. In the present study, we investigated the 5'-untranslated region (5'-UTR) of the mouse core clock gene, Per1, at the posttranscriptional level, particularly its translational regulation. The 5'-UTR of Per1 was found to promote its translation via an internal ribosomal entry site (IRES). We found that polypyrimidine tract-binding protein 1 (PTBP1) binds to the 5'-UTR of Per1 and positively regulates the IRES-mediated translation of Per1 without affecting the levels of Per1 mRNA. The reduction of PTBP1 level also decreased the endogenous levels of the PER1 protein but not of its mRNA. As for the oscillation of PER1 expression, the disruption of PTBP1 levels lowered the PER1 expression but not the phase of the oscillation. PTBP1 also changed the amplitudes of the mRNAs of other circadian clock genes, such as Cryptochrome 1 (Cry1) and Per3. Our results suggest that the PTBP1 is important for rhythmic translation of Per1 and it fine-tunes the overall circadian system.


Subject(s)
5' Untranslated Regions , Gene Expression Regulation , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Period Circadian Proteins/biosynthesis , Polypyrimidine Tract-Binding Protein/metabolism , Protein Biosynthesis , Animals , Cryptochromes/biosynthesis , Cryptochromes/genetics , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Mice , NIH 3T3 Cells , Period Circadian Proteins/genetics , Polypyrimidine Tract-Binding Protein/genetics
11.
Glia ; 67(9): 1667-1679, 2019 09.
Article in English | MEDLINE | ID: mdl-31050055

ABSTRACT

During postnatal neurodevelopment, excessive synapses must be eliminated by microglia to complete the establishment of neural circuits in the brain. The lack of synaptic regulation by microglia has been implicated in neurodevelopmental disorders such as autism, schizophrenia, and intellectual disability. Here we suggest that vaccinia-related kinase 2 (VRK2), which is expressed in microglia, may stimulate synaptic elimination by microglia. In VRK2-deficient mice (VRK2KO ), reduced numbers of presynaptic puncta within microglia were observed. Moreover, the numbers of presynaptic puncta and synapses were abnormally increased in VRK2KO mice by the second postnatal week. These differences did not persist into adulthood. Even though an increase in the number of synapses was normalized, adult VRK2KO mice showed behavioral defects in social behaviors, contextual fear memory, and spatial memory.


Subject(s)
Brain/enzymology , Brain/growth & development , Microglia/enzymology , Protein Serine-Threonine Kinases/metabolism , Synapses/enzymology , Animals , Brain/cytology , Cells, Cultured , Excitatory Postsynaptic Potentials/physiology , Fear/physiology , Humans , Male , Memory/physiology , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , Miniature Postsynaptic Potentials/physiology , Protein Serine-Threonine Kinases/genetics , Social Behavior , Tissue Culture Techniques
12.
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
13.
J Neurochem ; 147(5): 609-625, 2018 12.
Article in English | MEDLINE | ID: mdl-30062698

ABSTRACT

Vaccinia-related kinase 2 (VRK2) is a serine/threonine kinase that belongs to the casein kinase 1 family. VRK2 has long been known for its relationship with neurodegenerative disorders such as schizophrenia. However, the role of VRK2 and the substrates associated with it are unknown. Dysbindin is known as one of the strong risk factors for schizophrenia. The expression of dysbindin is indeed significantly reduced in schizophrenia patients. Moreover, dysbindin is involved in neurite outgrowth and regulation of NMDA receptor signaling. Here, we first identified dysbindin as a novel interacting protein of VRK2 through immunoprecipitation. We hypothesized that dysbindin is phosphorylated by VRK2 and further that this phosphorylation plays an important role in the function of dysbindin. We show that VRK2 phosphorylates Ser 297 and Ser 299 of dysbindin using in vitro kinase assay. In addition, we found that VRK2-mediated phosphorylation of dysbindin enhanced ubiquitination of dysbindin and consequently resulted in the decrease in its protein stability through western blotting. Over-expression of VRK2 in human neuroblastoma (SH-SY5Y) cells reduced neurite outgrowth induced by retinoic acid. Furthermore, a phosphomimetic mutant of dysbindin alleviated neurite outgrowth and affected surface expression of N-methyl-d-aspartate 2A, a subunit of NMDA receptor in mouse hippocampal neurons. Together, our work reveals the regulation of dysbindin by VRK2, providing the association of these two proteins, which are commonly implicated in schizophrenia. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.


Subject(s)
Dysbindin/physiology , Protein Serine-Threonine Kinases/physiology , Protein Stability , Animals , Cell Line , Dysbindin/genetics , Hippocampus/cytology , Hippocampus/metabolism , Humans , Mice , Mice, Inbred C57BL , Mutation/genetics , Mutation/physiology , Neurites/drug effects , Phosphorylation , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/pharmacology , Receptors, N-Methyl-D-Aspartate/biosynthesis , Tretinoin/pharmacology , Ubiquitination
14.
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
15.
Appl Microbiol Biotechnol ; 101(4): 1605-1614, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27858139

ABSTRACT

The functional features of Lactobacillus plantarum HAC01 (HAC01), isolated from fermented Korean kimchi, were studied with regard to the fat mass, immunometabolic biomarkers and dysbiosis in a diet-induced obesity (DIO) murine model. L. rhamnosus GG (LGG) served as reference strain and a PBS-treated group as control. The administration of L. plantarum HAC01 resulted in reduction of the mesenteric adipose depot, the conjunctive tissue closely associated with the gastrointestinal tract, where lipid oxidative gene expression was upregulated compared to the control group. Metagenome analysis of intestinal microbiota showed that both strains HAC01 and LGG influenced specific bacterial families such as the Lachnospiraceae and Ruminococcaceae rather than the phyla Firmicutes and Bacteroidetes as a whole. The relative abundance of the Lachnospiraceae (phylum Firmicutes) was significantly higher in both LAB-treated groups than in the control. Comparing the impact of the two Lactobacillus strains on microbial composition in the gut also suggests strain-specific effects. The study emphasises the need for deeper studies into functional specificity of a probiotic organism at the strain level. Alleviation of obesity-associated dysbiosis by modulation of the gut microbiota appears to be associated with "indicator" bacterial taxa such as the family Lachnospiraceae. This may provide further insight into mechanisms basic to the mode of probiotic action against obesity and associated dysbiosis.


Subject(s)
Adipose Tissue/metabolism , Gastrointestinal Microbiome/physiology , Lactobacillus plantarum/physiology , Obesity/metabolism , Obesity/microbiology , Animals , Diet, High-Fat/adverse effects , Mice , Obesity/etiology
16.
J Allergy Clin Immunol ; 137(2): 507-516.e8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26433560

ABSTRACT

BACKGROUND: The incidence of food allergies has increased dramatically during the last decade. Recently, probiotics have been studied for the prevention and treatment of allergic disease. OBJECTIVE: We examined whether Bifidobacterium longum KACC 91563 and Enterococcus faecalis KACC 91532 have the capacity to suppress food allergies. METHODS: B longum KACC 91563 and E faecalis KACC 91532 were administered to BALB/c wild-type mice, in which food allergy was induced by using ovalbumin and alum. Food allergy symptoms and various immune responses were assessed. RESULTS: B longum KACC 91563, but not E faecalis KACC 91532, alleviated food allergy symptoms. Extracellular vesicles of B longum KACC 91563 bound specifically to mast cells and induced apoptosis without affecting T-cell immune responses. Furthermore, injection of family 5 extracellular solute-binding protein, a main component of extracellular vesicles, into mice markedly reduced the occurrence of diarrhea in a mouse food allergy model. CONCLUSION: B longum KACC 91563 induces apoptosis of mast cells specifically and alleviates food allergy symptoms. Accordingly, B longum KACC 91563 and family 5 extracellular solute-binding protein exhibit potential as therapeutic approaches for food allergies.


Subject(s)
Bacterial Proteins/immunology , Bifidobacterium/immunology , Extracellular Vesicles/immunology , Food Hypersensitivity/immunology , Food Hypersensitivity/microbiology , Immunomodulation , Mast Cells/immunology , Animals , Apoptosis/immunology , Bacterial Proteins/metabolism , Bifidobacterium/metabolism , Cell Count , Cytokines/biosynthesis , Disease Models, Animal , Endocytosis/immunology , Extracellular Vesicles/metabolism , Food Hypersensitivity/metabolism , Intestine, Small/immunology , Intestine, Small/metabolism , Intestine, Small/microbiology , Intestine, Small/pathology , Mast Cells/metabolism , Mast Cells/microbiology , Mice , Probiotics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
17.
Biochim Biophys Acta ; 1853(7): 1738-48, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25899223

ABSTRACT

Vaccinia-related kinase 3 (VRK3) is known as a pseudokinase that is catalytically inactive due to changes in motifs that are essential for kinase activity. Although VRK3 has been regarded as a genuine pseudokinase from structural and biochemical studies, recent reports suggest that VRK3 acts as an active kinase as well as a signaling scaffold in cells. Here, we demonstrate that VRK3 phosphorylates the nuclear envelope protein barrier-to-autointegration factor (BAF) on Ser4. Interestingly, VRK3 kinase activity is dependent upon its N-terminal regulatory region, which is excluded from the determination of its crystal structure. Furthermore, the kinase activity of VRK3 is involved in the regulation of the cell cycle. VRK3 expression levels increase during interphase, whereas VRK1 is enriched in late G2 and early M phase. Ectopic expression of VRK3 induces the translocation of BAF from the nucleus to the cytoplasm. In addition, depletion of VRK3 decreases the population of proliferating cells. These data suggest that VRK3-mediated phosphorylation of BAF may facilitate DNA replication or gene expression by facilitating the dissociation of nuclear envelope proteins and chromatin during interphase.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Cycle , HEK293 Cells , HeLa Cells , Humans , Mice , Models, Biological , Nuclear Envelope/metabolism , Phosphorylation , Phosphoserine/metabolism , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Transport , Structure-Activity Relationship
18.
Nucleic Acids Res ; 42(6): 3590-606, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24423872

ABSTRACT

In the present study, we investigated the 3' untranslated region (UTR) of the mouse core clock gene cryptochrome 1 (Cry1) at the post-transcriptional level, particularly its translational regulation. Interestingly, the 3'UTR of Cry1 mRNA decreased its mRNA levels but increased protein amounts. The 3'UTR is widely known to function as a cis-acting element of mRNA degradation. The 3'UTR also provides a binding site for microRNA and mainly suppresses translation of target mRNAs. We found that AU-rich element RNA binding protein 1 (AUF1) directly binds to the Cry1 3'UTR and regulates translation of Cry1 mRNA. AUF1 interacted with eukaryotic translation initiation factor 3 subunit B and also directly associated with ribosomal protein S3 or ribosomal protein S14, resulting in translation of Cry1 mRNA in a 3'UTR-dependent manner. Expression of cytoplasmic AUF1 and binding of AUF1 to the Cry1 3'UTR were parallel to the circadian CRY1 protein profile. Our results suggest that the 3'UTR of Cry1 is important for its rhythmic translation, and AUF1 bound to the 3'UTR facilitates interaction with the 5' end of mRNA by interacting with translation initiation factors and recruiting the 40S ribosomal subunit to initiate translation of Cry1 mRNA.


Subject(s)
Circadian Rhythm/genetics , Cryptochromes/genetics , Gene Expression Regulation , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Protein Biosynthesis , RNA Stability , RNA, Messenger/metabolism , 3' Untranslated Regions , Animals , Cryptochromes/biosynthesis , Cryptochromes/metabolism , Eukaryotic Initiation Factors/metabolism , HEK293 Cells , Heterogeneous Nuclear Ribonucleoprotein D0 , Humans , Mice , NIH 3T3 Cells , Ribosomal Proteins/metabolism
19.
J Neurochem ; 132(6): 642-56, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25581122

ABSTRACT

The daily oscillations observed in most living organisms are endogenously generated with a period of 24 h, and the underlying structure of periodic oscillation is an autoregulatory transcription-translation feedback loop. The mechanisms of untranslated region (UTR)-mediated post-transcriptional regulation (e.g., mRNA degradation and internal ribosomal entry site (IRES)-mediated translation) have been suggested to fine-tune the expression of clock genes. Mouse Period3 (mPer3) is one of the paralogs of Period gene and its function is important in peripheral clocks and sleep physiology. mPer3 mRNA displays a circadian oscillation as well as a circadian phase-dependent stability, while the stability regulators still remain unknown. In this study, we identify three proteins - heterogeneous nuclear ribonucleoprotein (hnRNP) K, polypyrimidine tract-binding protein (PTB), and hnRNP D - that bind to mPer3 mRNA 3'-UTR. We show that hnRNP K is a stabilizer that increases the amplitude of circadian mPer3 mRNA oscillation and hnRNP D is a destabilizer that decreases it, while PTB exhibits no effect on mPer3 mRNA expression. Our experiments describe their cytoplasmic roles for the mRNA stability regulation and the circadian amplitude formation. Moreover, our mathematical model suggests a mechanism through which post-transcriptional mRNA stability modulation provides not only the flexibility of oscillation amplitude, but also the robustness of the period and the phase for circadian mPer3 expression. Mouse Period3 (mPer3) is one of well-known clock genes. We identified three 3'-UTR-binding proteins that modulate the mRNA stability, and they influenced to the amplitude of circadian mPer3 mRNA oscillation. Our mathematical model not only showed the relationship between mRNA stability and its oscillation profile but provided the molecular mechanism for the robustness of the period and the phase in circadian oscillation. hnK, heterogeneous nuclear ribonucleoprotein (hnRNP) K; hnD, hnRNP D; PTB, polypyrimidine tract-binding protein.


Subject(s)
Circadian Rhythm/physiology , Period Circadian Proteins/biosynthesis , RNA, Messenger/physiology , Animals , Cell Line, Tumor , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , Protein Stability , RNA Processing, Post-Transcriptional/physiology
20.
J Pharmacol Exp Ther ; 352(1): 175-84, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25369797

ABSTRACT

To date, many anticancer drugs have been developed by directly or indirectly targeting microtubules, which are involved in cell division. Although this approach has yielded many anticancer drugs, these drugs produce undesirable side effects. An alternative strategy is needed, and targeting mitotic exit may be one alternative approach. Localization of phosphorylated barrier-to-autointegration factor (BAF) to the chromosomal core region is essential for nuclear envelope compartment relocalization. In this study, we isolated brazilin from Caesalpinia sappan Leguminosae and demonstrated that it inhibited BAF phosphorylation in vitro and in vivo. Moreover, we demonstrated direct binding between brazilin and BAF. The inhibition of BAF phosphorylation induced abnormal nuclear envelope reassembly and cell death, indicating that perturbation of nuclear envelope reassembly could be a novel approach to anticancer therapy. We propose that brazilin isolated from C. sappan may be a new anticancer drug candidate that induces cell death by inhibiting vaccinia-related kinase 1-mediated BAF phosphorylation.


Subject(s)
Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Benzopyrans/isolation & purification , Benzopyrans/pharmacology , Caesalpinia/chemistry , DNA-Binding Proteins/metabolism , Nuclear Envelope/drug effects , Nuclear Proteins/metabolism , Animals , Antineoplastic Agents/metabolism , Benzopyrans/metabolism , Cell Death/drug effects , Drug Evaluation, Preclinical , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Nuclear Envelope/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Telophase/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL