Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37961595

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder caused by a CAG repeat expansion in the first exon of the HTT gene encoding huntingtin. Prior reports have established a correlation between CAG expanded HTT and altered gene expression. However, the mechanisms leading to disruption of RNA processing in HD remain unclear. Here, our analysis of the reported HTT protein interactome identifies interactions with known RNA-binding proteins (RBPs). Total, long-read sequencing and targeted RASL-seq of RNAs from cortex and striatum of the HD mouse model R6/2 reveals increased exon skipping which is confirmed in Q150 and Q175 knock-in mice and in HD human brain. We identify the RBP TDP-43 and the N6-methyladenosine (m6A) writer protein methyltransferase 3 (METTL3) to be upstream regulators of exon skipping in HD. Along with this novel mechanistic insight, we observe decreased nuclear localization of TDP-43 and cytoplasmic accumulation of phosphorylated TDP-43 in HD mice and human brain. In addition, TDP-43 co-localizes with HTT in human HD brain forming novel nuclear aggregate-like bodies distinct from mutant HTT inclusions or previously observed TDP-43 pathologies. Binding of TDP-43 onto RNAs encoding HD-associated differentially expressed and aberrantly spliced genes is decreased. Finally, m6A RNA modification is reduced on RNAs abnormally expressed in striatum from HD R6/2 mouse brain, including at clustered sites adjacent to TDP-43 binding sites. Our evidence supports TDP-43 loss of function coupled with altered m6A modification as a novel mechanism underlying alternative splicing/unannotated exon usage in HD and highlights the critical nature of TDP-43 function across multiple neurodegenerative diseases.

2.
J Biol Chem ; 299(9): 105175, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37599001

ABSTRACT

N6-adenosine methylation (m6A) is the most abundant mRNA modification that controls gene expression through diverse mechanisms. Accordingly, m6A-dependent regulation of oncogenes and tumor suppressors contributes to tumor development. However, the role of m6A-mediated gene regulation upon drug treatment or resistance is poorly understood. Here, we report that m6A modification of mitogen-activated protein kinase 13 (MAPK13) mRNA determines the sensitivity of cancer cells to the mechanistic target of rapamycin complex 1 (mTORC1)-targeting agent rapamycin. mTORC1 induces m6A modification of MAPK13 mRNA at its 3' untranslated region through the methyltransferase-like 3 (METTL3)-METTL14-Wilms' tumor 1-associating protein(WTAP) methyltransferase complex, facilitating its mRNA degradation via an m6A reader protein YTH domain family protein 2. Rapamycin blunts this process and stabilizes MAPK13. On the other hand, genetic or pharmacological inhibition of MAPK13 enhances rapamycin's anticancer effects, which suggests that MAPK13 confers a progrowth signal upon rapamycin treatment, thereby limiting rapamycin efficacy. Together, our data indicate that rapamycin-mediated MAPK13 mRNA stabilization underlies drug resistance, and it should be considered as a promising therapeutic target to sensitize cancer cells to rapamycin.

3.
Mol Cells ; 45(7): 435-443, 2022 Jul 31.
Article in English | MEDLINE | ID: mdl-35748227

ABSTRACT

In response to environmental changes, signaling pathways rewire gene expression programs through transcription factors. Epigenetic modification of the transcribed RNA can be another layer of gene expression regulation. N6-adenosine methylation (m6A) is one of the most common modifications on mRNA. It is a reversible chemical mark catalyzed by the enzymes that deposit and remove methyl groups. m6A recruits effector proteins that determine the fate of mRNAs through changes in splicing, cellular localization, stability, and translation efficiency. Emerging evidence shows that key signal transduction pathways including TGFß (transforming growth factor-ß), ERK (extracellular signal-regulated kinase), and mTORC1 (mechanistic target of rapamycin complex 1) regulate downstream gene expression through m6A processing. Conversely, m6A can modulate the activity of signal transduction networks via m6A modification of signaling pathway genes or by acting as a ligand for receptors. In this review, we discuss the current understanding of the crosstalk between m6A and signaling pathways and its implication for biological systems.


Subject(s)
Adenosine , Signal Transduction , Adenosine/genetics , Adenosine/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Methylation , RNA, Messenger/genetics , Transforming Growth Factor beta/metabolism
4.
ACS Chem Neurosci ; 13(6): 806-817, 2022 03 16.
Article in English | MEDLINE | ID: mdl-35230076

ABSTRACT

α-Synuclein accumulation is implicated in the pathogenesis of neurodegenerative diseases, including Parkinson's disease (PD). Previously, we reported that Fas-associated factor 1 (FAF1), which plays a role in PD pathogenesis, potentiates α-synuclein accumulation through autophagy impairment in dopaminergic neurons. In this study, we show that KM-819, a FAF1-targeting compound, which has completed phase I clinical trials, interferes with α-synuclein accumulation in the mouse brain, as well as in human neuronal cells (SH-SY5Ys). KM-819 suppressed the accumulation of monomeric, oligomeric, and aggregated forms of α-synuclein in neuronal cells. Furthermore, KM-819 restored the turnover rate of α-synuclein in FAF1-overexpressing SH-SY5Y cells, implicating KM-819-mediated reconstitution of the α-synuclein degradative pathway. In addition, KM-819 reconstituted autophagic flux in FAF1-transfected SH-SY5Y cells, also suppressing α-synuclein-induced mitochondrial dysfunction. Moreover, oral administration of KM-819 also interfered with α-synuclein accumulation in the midbrain of mice overexpressing FAF1 via an adeno-associated virus system. Consistently, KM-819 reduced α-synuclein accumulation in both the hippocampus and the midbrain of human A53T α-synuclein transgenic mice. Collectively, these data imply that KM-819 may have therapeutic potential for patients with PD.


Subject(s)
Parkinson Disease , alpha-Synuclein , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Autophagy/physiology , Brain/metabolism , Disease Models, Animal , Dopaminergic Neurons/metabolism , Humans , Mice , Mice, Transgenic , Parkinson Disease/metabolism , alpha-Synuclein/metabolism
5.
Biomed Pharmacother ; 133: 111041, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33378949

ABSTRACT

Poly (ADP-ribose) polymerase 1 (PARP1)-dependent cell death in the retinal pigment epithelium (RPE) is implicated in dry age-related macular degeneration (AMD). Although PARP1 inhibitors are available for treating dry AMD, their delivery route is not ideal for patients. The aim of this study was to test the efficacy of a novel PARP1-inhibitory compound (PIC) in vitro and in vivo. This study presents PIC, a novel small molecule, with superior efficacy to PARP1 inhibitors in the market. PIC demonstrated a distinctive inhibitory profile against PARP isotypes than the FDA-approved PARP1 inhibitors. PIC inhibited PARP1 activation at an IC50 of 0.41 ± 0.15 nM in an enzyme-based assay in vitro and at IC50 and EC50 in ARPE-19 cells of 0.11 ± 0.02 nM and 0.22 ± 0.02 nM, respectively, upon H2O2 insult. PIC also moderated mitochondrial fission and depolarization and maintained cellular energy levels under oxidative stress in ARPE-19 cells. Furthermore, PIC demonstrated good corneal penetration in a rat model, presenting PIC as a promising candidate for eye drop therapeutics for dry AMD. When PIC was administered as an eye drop formulation, RPE morphology was preserved, maintaining the thickness of the outer nuclear layers under sodium iodate (SI) treatment in rats. In SI-treated rabbits, eye drop administration of PIC also retained the structural and functional integrity when analyzed using funduscopy and electroretinogram. Collectively, our data portray PIC as an attractive treatment measure for dry AMD.


Subject(s)
Macular Degeneration/drug therapy , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Retinal Pigment Epithelium/drug effects , Administration, Ophthalmic , Animals , Antioxidants/pharmacology , Cell Line , Disease Models, Animal , Humans , Iodates , Macular Degeneration/chemically induced , Macular Degeneration/enzymology , Macular Degeneration/pathology , Male , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Ocular Absorption , Ophthalmic Solutions , Oxidative Stress/drug effects , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Rabbits , Rats, Sprague-Dawley , Retinal Pigment Epithelium/enzymology , Retinal Pigment Epithelium/pathology
6.
Mol Cells ; 43(7): 632-644, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32732457

ABSTRACT

The molecular mechanism underlying autophagy impairment in the retinal pigment epithelium (RPE) in dry age-related macular degeneration (AMD) is not yet clear. Based on the causative role of poly(ADP-ribose) polymerase 1 (PARP1) in RPE necrosis, this study examined whether PARP1 is involved in the autophagy impairment observed during dry AMD pathogenesis. We found that autophagy was downregulated following H2O2-induced PARP1 activation in ARPE-19 cells and olaparib, PARP1 inhibitor, preserved the autophagy process upon H2O2 exposure in ARPE-19 cells. These findings imply that PARP1 participates in the autophagy impairment upon oxidative stress in ARPE-19 cells. Furthermore, PARP1 inhibited autolysosome formation but did not affect autophagosome formation in H2O2-exposed ARPE-19 cells, demonstrating that PARP1 is responsible for impairment of late-stage autophagy in particular. Because PARP1 consumes NAD+ while exerting its catalytic activity, we investigated whether PARP1 impedes autophagy mediated by sirtuin1 (SIRT1), which uses NAD+ as its cofactor. A NAD+ precursor restored autophagy and protected mitochondria in ARPE-19 cells by preserving SIRT1 activity upon H2O2. Moreover, olaparib failed to restore autophagy in SIRT1-depleted ARPE-19 cells, indicating that PARP1 inhibits autophagy through SIRT1 inhibition. Next, we further examined whether PARP1-induced autophagy impairment occurs in the retinas of dry AMD model mice. Histological analyses revealed that olaparib treatment protected mouse retinas against sodium iodate (SI) insult, but not in retinas cotreated with SI and wortmannin, an autophagy inhibitor. Collectively, our data demonstrate that PARP1-dependent inhibition of SIRT1 activity impedes autophagic survival of RPE cells, leading to retinal degeneration during dry AMD pathogenesis.


Subject(s)
Autophagy/genetics , Macular Degeneration/metabolism , Oxidative Stress/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Retinal Pigment Epithelium/metabolism , Sirtuin 1/metabolism , Animals , Autophagy/drug effects , Disease Models, Animal , Humans , Hydrogen Peroxide/toxicity , Iodates/toxicity , Lysosomes/drug effects , Lysosomes/genetics , Lysosomes/metabolism , Macular Degeneration/genetics , Macular Degeneration/pathology , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , NAD/metabolism , Oxidative Stress/drug effects , Phthalazines/pharmacology , Piperazines/pharmacology , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Retina/drug effects , Retina/metabolism , Retina/pathology , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , Wortmannin/pharmacology
7.
Exp Eye Res ; 180: 8-17, 2019 03.
Article in English | MEDLINE | ID: mdl-30500363

ABSTRACT

Receptor interacting protein kinase 1 (RIPK1) plays a key role in necroptosis, which is a type of programmed necrosis that is involved in ocular diseases, including glaucoma and dry age-related macular degeneration (AMD). We previously introduced RIPK1-inhibitory compound (RIC), which has biochemical characteristics and a mode of action that are distinct from those of the prototype RIPK1 inhibitor necrostatin-1. The intraperitoneal administration of RIC exerts a protective effect on retinal ganglion cells against a glaucomatous insult. In this study, we examined the protective effect of RIC on retinal pigment epithelium (RPE) against sodium iodate (SI) insult, which is associated with dry AMD pathogenesis. The eye drop administration of RIC that reached on the retina prevented RPE loss in SI-induced retinal degeneration. RIC consistently demonstrated retinal protection in the funduscopy and electroretinogram analyses in SI-injected rabbits and iodoacetic acid-treated mini-pigs. Moreover, the in vivo protective effects of RIC were superior to those of ACU-4429 and doxycycline, which are other medications investigated in clinical trials for the treatment of dry AMD, and RIC did not induce retinal toxicity following topical administration in rats. Collectively, RIC displayed excellent retinal penetration and prevented retinal degeneration in the pathogenesis of dry AMD with a high in vivo efficacy.


Subject(s)
Disease Models, Animal , Geographic Atrophy/prevention & control , Protective Agents/therapeutic use , Receptor-Interacting Protein Serine-Threonine Kinases/therapeutic use , Retinal Ganglion Cells/drug effects , Administration, Ophthalmic , Animals , Electroretinography , Geographic Atrophy/chemically induced , Geographic Atrophy/pathology , Iodates/toxicity , Male , Ophthalmoscopy , Phenyl Ethers/therapeutic use , Propanolamines/therapeutic use , Rabbits , Rats , Rats, Sprague-Dawley , Retinal Degeneration/prevention & control
8.
Biochim Biophys Acta Mol Cell Res ; 1865(1): 132-141, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28993228

ABSTRACT

Regulated necrosis occurs in various pathophysiological conditions under oxidative stress. Here, we report that receptor-interacting protein kinase 1 (RIPK1), a key player in one type of regulated necrosis (necroptosis), also participates in another type of poly (ADP-ribose) polymerase 1 (PARP1)-dependent regulated necrosis (parthanatos). Various biological signatures of parthanatos were significantly attenuated in Ripk1-/- mouse embryonic fibroblasts, including PARylation, nuclear translocation of apoptosis-inducing factor, and PARP1-dependent cell death under H2O2 exposure. Hence, we investigated whether RIPK1 regulates the activity of PARP1. RIPK1 activated PARP1 via an interaction with the catalytic domain of PARP1 in the nucleus. Of note, both wild type and kinase-dead mutant RIPK1 induced PARP1 activation and led to PARP1-mediated cell death upon H2O2 insult, demonstrating the kinase-independent regulation of RIPK1 in PARP1 activation. Collectively, our results demonstrate the existence of a kinase-independent role of nuclear RIPK1 in the regulation of PARP1.


Subject(s)
Necrosis/genetics , Oxidative Stress/physiology , Poly (ADP-Ribose) Polymerase-1/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Animals , Apoptosis/genetics , Cell Nucleus/metabolism , Cells, Cultured , HEK293 Cells , Humans , Mice , Necrosis/metabolism , Phosphorylation , Protein Binding , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics
9.
Exp Cell Res ; 359(1): 30-38, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28803066

ABSTRACT

In glaucoma, retinal ganglion cells (RGCs) are exposed to ischemic stress with elevation of the intraocular pressure and are subsequently lost. Necroptosis, a type of regulated necrosis, is known to play a pivotal role in this loss. We observed that receptor-interacting protein kinase 1 (RIPK1), the key player of necroptosis, was activated by diverse ischemic stresses, including TCZ, chemical hypoxia (CH), and oxygen glucose deprivation (OGD). In this study, we introduce a RIPK1-inhibitory compound (RIC) with a novel scaffold. RIC inhibited downstream events following RIPK1 activation, including necrosome formation and mitochondrial dysfunction in RGC5 cells. Moreover, RIC protected RGCs against ischemic injury in the rat glaucoma model, which was induced by acute high intraocular pressure. However, RIC displayed biochemical characteristics that are distinct from those of previous RIPK1 inhibitors (necrostatin-1; Nec-1 and Compound 27; Cpd27). RIC protected RGCs against OGD insult, while Nec-1 and Cpd27 did not. Conversely, Nec-1 and Cpd27 protected RGCs from TNF-stimulated death, while RIC failed to inhibit the death of RGCs. This implies that RIPK1 activates alternative pathways depending on the context of the ischemic insults.


Subject(s)
Glaucoma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Retinal Degeneration/drug therapy , Retinal Degeneration/prevention & control , Animals , Apoptosis/drug effects , Cell Hypoxia/drug effects , Cells, Cultured , Cycloheximide , Disease Models, Animal , Glaucoma/complications , Glaucoma/pathology , Glucose/deficiency , HT29 Cells , Humans , Injections, Intraperitoneal , Ischemia/complications , Ischemia/pathology , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Necrosis , Neuroprotection/drug effects , Oligopeptides , Oxygen , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Rats, Sprague-Dawley , Receptor-Interacting Protein Serine-Threonine Kinases , Retinal Degeneration/complications , Retinal Degeneration/pathology , Retinal Neurons/drug effects , Retinal Neurons/metabolism , Retinal Neurons/pathology , Tumor Necrosis Factor-alpha
10.
Cell Death Dis ; 8(1): e2526, 2017 01 05.
Article in English | MEDLINE | ID: mdl-28055012

ABSTRACT

Cell death of retinal pigment epithelium (RPE) is characterized as an essential late-stage phenomenon of dry age-related macular degeneration (AMD). The aim of this study was to elucidate the molecular mechanism underlying RPE cell death after exposure to oxidative stress, which occurs often because of the anatomical location of RPE cells. ARPE-19, an established RPE cell line, exhibited necrotic features involving poly (ADP-ribose) polymerase-1 (PARP-1) activation in response to hydrogen peroxide (H2O2). ARPE-19 cells were resistant to H2O2 when PARP-1 was depleted using siRNA or inhibited by a pharmacological inhibitor of PARP-1, olaparib. Our data suggest a causal relationship between PARP-1 activation and ARPE-19 cell death in response to H2O2. Next, we investigated downstream molecular events in PARP-1 activation. Increased mitochondrial depolarization, mitochondrial fission and alterations of the cellular energy dynamics with reduced NAD+ and ATP were observed in H2O2-treated ARPE-19 cells. H2O2-triggered mitochondrial dysfunction was inhibited by olaparib. Nevertheless, translocation of apoptosis-inducing factor (AIF), a biochemical signature for PARP-1-dependent cell death (parthanatos), was not observed in our study. Moreover, the depletion of AIF did not affect the amplitude of cell death, demonstrating the lack of a role for AIF in the death of ARPE-19 cells in response to H2O2. This feature distinguishes the type of death observed in this study from canonical parthanatos. Next, we examined the in vivo role of PARP-1 in a dry AMD animal model system. Histological analysis of the outer nuclear layer in the mouse retina revealed protection against sodium iodate (SI) following treatment with olaparib. Moreover, retina fundus and electroretinograms also confirmed such a protective effect in the SI-treated rabbit. Collectively, we report that AIF-independent PARP-1-dependent necrosis constitutes a major mechanism of RPE cell death leading to retinal degeneration in dry AMD.


Subject(s)
Apoptosis Inducing Factor/genetics , Geographic Atrophy/genetics , Macular Degeneration/genetics , Necrosis/genetics , Poly (ADP-Ribose) Polymerase-1/genetics , Animals , Cell Death/genetics , Disease Models, Animal , Geographic Atrophy/drug therapy , Geographic Atrophy/pathology , Humans , Hydrogen Peroxide/toxicity , Macular Degeneration/drug therapy , Macular Degeneration/pathology , Mice , Mitochondrial Dynamics/drug effects , Necrosis/drug therapy , Necrosis/pathology , Phthalazines/administration & dosage , Piperazines/administration & dosage , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...