Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 133(16)2023 08 15.
Article in English | MEDLINE | ID: mdl-37384409

ABSTRACT

Protein aggregation is a hallmark of many neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS). Although mutations in TARDBP, encoding transactive response DNA-binding protein 43 kDa (TDP-43), account for less than 1% of all ALS cases, TDP-43-positive aggregates are present in nearly all ALS patients, including patients with sporadic ALS (sALS) or carrying other familial ALS-causing (fALS-causing) mutations. Interestingly, TDP-43 inclusions are also present in subsets of patients with frontotemporal dementia, Alzheimer's disease, and Parkinson's disease; therefore, methods of activating intracellular protein quality control machinery capable of clearing toxic cytoplasmic TDP-43 species may alleviate disease-related phenotypes. Here, we identify a function of nemo-like kinase (Nlk) as a negative regulator of lysosome biogenesis. Genetic or pharmacological reduction of Nlk increased lysosome formation and improved clearance of aggregated TDP-43. Furthermore, Nlk reduction ameliorated pathological, behavioral, and life span deficits in 2 distinct mouse models of TDP-43 proteinopathy. Because many toxic proteins can be cleared through the autophagy/lysosome pathway, targeted reduction of Nlk represents a potential approach to therapy development for multiple neurodegenerative disorders.


Subject(s)
Amyotrophic Lateral Sclerosis , Neurodegenerative Diseases , Animals , Mice , Amyotrophic Lateral Sclerosis/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Lysosomes/metabolism , Neurodegenerative Diseases/genetics , Humans
2.
ACS Sens ; 7(4): 1068-1074, 2022 04 22.
Article in English | MEDLINE | ID: mdl-35353484

ABSTRACT

Cancer cells undergo unscheduled proliferation resulting from dysregulation of the cell cycle, and hence, evaluation in tumor is of keen interest to examine the invasiveness and recurrence of cancer in the lesion. Molecular probes capable of discriminating actively growing tumor from resting ones remain unexplored despite their vast importance. Here, we describe a novel strategy to visualize invasive areas in tumor with a fluorescence probe that implements synergistic fluorescence response toward the slightly acidic environment of tumor and an ATP-abundant nature of actively growing cells. The probe has been designed for ultrafast detection of ATP with high specificity. We demonstrate its utility in visualizing invasive areas in tumor by distinguishing basal cell carcinomas and squamous cell carcinomas at their early stages by two-photon microscopy.


Subject(s)
Carcinoma, Squamous Cell , Skin Neoplasms , Adenosine Triphosphate , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Humans , Protons , Skin/metabolism , Skin Neoplasms/diagnosis , Skin Neoplasms/pathology
3.
JCI Insight ; 6(22)2021 11 22.
Article in English | MEDLINE | ID: mdl-34618685

ABSTRACT

Genetic variants in Granulin (GRN), which encodes the secreted glycoprotein progranulin (PGRN), are associated with several neurodegenerative diseases, including frontotemporal lobar degeneration, neuronal ceroid lipofuscinosis, and Alzheimer's disease. These genetic alterations manifest in pathological changes due to a reduction of PGRN expression; therefore, identifying factors that can modulate PGRN levels in vivo would enhance our understanding of PGRN in neurodegeneration and could reveal novel potential therapeutic targets. Here, we report that modulation of the endocytosis/lysosomal pathway via reduction of Nemo-like kinase (Nlk) in microglia, but not in neurons, can alter total brain Pgrn levels in mice. We demonstrate that Nlk reduction promotes Pgrn degradation by enhancing its trafficking through the endocytosis/lysosomal pathway, specifically in microglia. Furthermore, genetic interaction studies in mice showed that Nlk heterozygosity in Grn haploinsufficient mice further reduces Pgrn levels and induces neuropathological phenotypes associated with PGRN deficiency. Our results reveal a mechanism for Pgrn level regulation in the brain through the active catabolism by microglia and provide insights into the pathophysiology of PGRN-associated diseases.


Subject(s)
Endocytosis/genetics , Lysosomes/metabolism , Microglia/metabolism , Progranulins/metabolism , Animals , Disease Models, Animal , Humans , Mice
4.
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
5.
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.

6.
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
7.
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
8.
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
9.
PLoS One ; 11(7): e0159018, 2016.
Article in English | MEDLINE | ID: mdl-27392095

ABSTRACT

Precise regulation of gene expression is especially important for circadian timekeeping which is maintained by the proper oscillation of the mRNA and protein of clock genes and clock-controlled genes. As a main component of the core negative arm feedback loops in the circadian clock, the Cry1 gene contributes to the maintenance of behavioral and molecular rhythmicity. Despite the central role of Cry1, the molecular mechanisms regulating expression levels of Cry1 mRNA and protein are not well defined. In particular, the post-transcriptional regulation of Cry1 mRNA fate decisions is unclear. Here, we demonstrate that hnRNP Q binds to mCry1 mRNA via the 5'UTR. Furthermore, hnRNP Q inhibits the translation of mCry1 mRNA, leading to altered rhythmicity in the mCRY1 protein profile.


Subject(s)
5' Untranslated Regions/physiology , Cryptochromes/biosynthesis , Gene Expression Regulation/physiology , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Protein Biosynthesis/physiology , Animals , Cryptochromes/genetics , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Mice , NIH 3T3 Cells
10.
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
11.
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
12.
Chem Commun (Camb) ; 52(1): 124-7, 2016 Jan 04.
Article in English | MEDLINE | ID: mdl-26503088

ABSTRACT

The selective detection of zinc ions in lysosomes over that in cytosol is achieved with a fluorescent probe, which enabled the fluorescence imaging of endogenous zinc ions in lysosomes of NIH 3T3 cells as well as mouse hippocampal tissues by two-photon microscopy under excitation at 900 nm.


Subject(s)
Fluorescent Dyes/chemistry , Hippocampus/chemistry , Lysosomes/chemistry , Zinc/analysis , Animals , Cations, Divalent/analysis , Mice , Microscopy, Fluorescence , NIH 3T3 Cells , Optical Imaging
13.
J Pineal Res ; 59(4): 518-29, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26444903

ABSTRACT

Rhythmic arylalkylamine N-acetyltransferase (AANAT) synthesis is a prominent circadian-controlled response that occurs in most mammals. AANAT is the core enzyme in melatonin production; because melatonin participates in many physiological processes, the regulation of AANAT is an important research topic. In this study, we focused on the role of heterogeneous ribonucleoprotein R (hnRNP R) in the translation of AANAT. A novel RNA-binding protein hnRNP R widely interacted with the 5' untranslated region (UTR) of AANAT mRNA and contributed to translation through an internal ribosomal entry site (IRES). Fine-tuning of AANAT protein synthesis occurred in response to knockdown and overexpression of hnRNP R. Nocturnal elevation of AANAT protein was dependent on the rhythmic changes of hnRNP R, whose levels are elevated in the pineal gland during nighttime. Increases in hnRNP R additionally improved AANAT production in rat pinealocytes under norepinephrine (NE) treatment. These results suggest that cap-independent translation of AANAT mRNA plays a role in the rhythmic synthesis of melatonin through the recruitment of translational machinery to hnRNP R-bound AANAT mRNA.


Subject(s)
Melatonin/metabolism , Animals , Arylalkylamine N-Acetyltransferase/metabolism , Blotting, Western , Cell Line , Humans , Immunohistochemistry , Internal Ribosome Entry Sites/genetics , Norepinephrine/pharmacology , Rats , Rats, Sprague-Dawley , Ribonucleoproteins/metabolism
14.
PLoS One ; 9(10): e109655, 2014.
Article in English | MEDLINE | ID: mdl-25310002

ABSTRACT

Uncontrolled proliferation, a major feature of cancer cells, is often triggered by the malfunction of cell cycle regulators such as protein kinases. Recently, cell cycle-related protein kinases have become attractive targets for anti-cancer therapy, because they play fundamental roles in cellular proliferation. However, the protein kinase-targeted drugs that have been developed so far do not show impressive clinical results and also display severe side effects; therefore, there is undoubtedly a need to investigate new drugs targeting other protein kinases that are critical in cell cycle progression. Vaccinia-related kinase 1 (VRK1) is a mitotic kinase that functions in cell cycle regulation by phosphorylating cell cycle-related substrates such as barrier-to-autointegration factor (BAF), histone H3, and the cAMP response element (CRE)-binding protein (CREB). In our study, we identified luteolin as the inhibitor of VRK1 by screening a small-molecule natural compound library. Here, we evaluated the efficacy of luteolin as a VRK1-targeted inhibitor for developing an effective anti-cancer strategy. We confirmed that luteolin significantly reduces VRK1-mediated phosphorylation of the cell cycle-related substrates BAF and histone H3, and directly interacts with the catalytic domain of VRK1. In addition, luteolin regulates cell cycle progression by modulating VRK1 activity, leading to the suppression of cancer cell proliferation and the induction of apoptosis. Therefore, our study suggests that luteolin-induced VRK1 inhibition may contribute to establish a novel cell cycle-targeted strategy for anti-cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Luteolin/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Antineoplastic Agents/chemistry , Catalytic Domain/drug effects , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Luteolin/chemistry , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/chemistry
15.
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
16.
PLoS One ; 8(11): e81218, 2013.
Article in English | MEDLINE | ID: mdl-24278397

ABSTRACT

Recently, it was found that microglia regulated synaptic remodeling of the developing brain, but their mechanisms have not been well understood. In this study, the action of microglia on neuronal synapse formation was investigated, and the primary target of microglial processes was discovered. When the developing microglia were applied to cultured hippocampal neurons without direct contact, the numbers of dendritic spines and excitatory and inhibitory synapses significantly increased. In order to find out the main factor for synaptic formation, the effects of cytokines released from microglia were examined. When recombinant proteins of cytokines were applied to neuronal culture media, interleukin 10 increased the numbers of dendritic spines in addition to excitatory and inhibitory synapses. Interestingly, without external stimuli, the amount of interleukin 10 released from the intact microglia appeared to be sufficient for the induction of synaptic formation. The neutralizing antibodies of interleukin 10 receptors attenuated the induction of the synaptic formation by microglia. The expression of interleukin 10 receptor was newly found in the hippocampal neurons of early developmental stage. When interleukin 10 receptors on the hippocampal neurons were knocked down with specific shRNA, the induction of synaptic formation by microglia and interleukin 10 disappeared. Pretreatment with lipopolysaccharide inhibited microglia from inducing synaptic formation, and interleukin 1ß antagonized the induction of synaptic formation by interleukin 10. In conclusion, the developing microglia regulated synaptic functions and neuronal development through the interactions of the interleukin 10 released from the microglia with interleukin 10 receptors expressed on the hippocampal neurons.


Subject(s)
Interleukin-10/metabolism , Microglia/metabolism , Neuroeffector Junction/metabolism , Animals , Animals, Newborn , Gene Expression , Gene Knockdown Techniques , Hippocampus/cytology , Hippocampus/metabolism , Interleukin-10/pharmacology , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Neuroeffector Junction/drug effects , Pyramidal Cells/metabolism , Rats , Receptors, Interleukin-10/genetics , Receptors, Interleukin-10/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...