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










Publication year range
1.
Aging Biol ; 12024.
Article in English | MEDLINE | ID: mdl-38500536

ABSTRACT

There is considerable interest in whether sensory deficiency is associated with the development of Alzheimer's disease (AD). Notably, the relationship between hearing impairment and AD is of high relevance but still poorly understood. In this study, we found early-onset hearing loss in two AD mouse models, 3xTgAD and 3xTgAD/Polß+/-. The 3xTgAD/Polß+/- mouse is DNA repair deficient and has more humanized AD features than the 3xTgAD. Both AD mouse models showed increased auditory brainstem response (ABR) thresholds between 16 and 32 kHz at 4 weeks of age, much earlier than any AD cognitive and behavioral changes. The ABR thresholds were significantly higher in 3xTgAD/Polß+/- mice than in 3xTgAD mice at 16 kHz, and distortion product otoacoustic emission signals were reduced, indicating that DNA damage may be a factor underlying early hearing impairment in AD. Poly ADP-ribosylation and protein expression levels of DNA damage markers increased significantly in the cochlea of the AD mice but not in the adjacent auditory cortex. Phosphoglycerate mutase 2 levels and the number of synaptic ribbons in the presynaptic zones of inner hair cells were decreased in the cochlea of the AD mice. Furthermore, the activity of sirtuin 3 was downregulated in the cochlea of these mice, indicative of impaired mitochondrial function. Taken together, these findings provide new insights into potential mechanisms for hearing dysfunction in AD and suggest that DNA damage in the cochlea might contribute to the development of early hearing loss in AD.

2.
Aging Cell ; 22(9): e13909, 2023 09.
Article in English | MEDLINE | ID: mdl-37395319

ABSTRACT

Age-related hearing loss (ARHL) is the most common sensory disability associated with human aging. Yet, there are no approved measures for preventing or treating this debilitating condition. With its slow progression, continuous and safe approaches are critical for ARHL treatment. Nicotinamide Riboside (NR), a NAD+ precursor, is well tolerated even for long-term use and is already shown effective in various disease models including Alzheimer's and Parkinson's disease. It has also been beneficial against noise-induced hearing loss and in hearing loss associated with premature aging. However, its beneficial impact on ARHL is not known. Using two different wild-type mouse strains, we show that long-term NR administration prevents the progression of ARHL. Through transcriptomic and biochemical analysis, we find that NR administration restores age-associated reduction in cochlear NAD+ levels, upregulates biological pathways associated with synaptic transmission and PPAR signaling, and reduces the number of orphan ribbon synapses between afferent auditory neurons and inner hair cells. We also find that NR targets a novel pathway of lipid droplets in the cochlea by inducing the expression of CIDEC and PLIN1 proteins that are downstream of PPAR signaling and are key for lipid droplet growth. Taken together, our results demonstrate the therapeutic potential of NR treatment for ARHL and provide novel insights into its mechanism of action.


Subject(s)
NAD , Presbycusis , Humans , Animals , Mice , Peroxisome Proliferator-Activated Receptors , Presbycusis/drug therapy , Presbycusis/prevention & control , Cochlea , Dietary Supplements
3.
Ann Biomed Eng ; 50(12): 1762-1770, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36369597

ABSTRACT

Mitochondria are highly dynamic multifaceted organelles with various functions including cellular energy metabolism, reactive oxygen species (ROS) generation, calcium homeostasis, and apoptosis. Because of these diverse functions, mitochondria are key regulators of cell survival and death, and their dysfunction is implicated in numerous diseases, particularly neurodegenerative disorders such as Alzheimer's Disease, Parkinson's Disease, and Huntington's Disease. One of the most common neurodegenerative disorders is sensorineural hearing loss (SNHL). SNHL primarily originates from the degenerative changes in the cochlea, which is the auditory portion of the inner ear. Many cochlear cells contain an abundance of mitochondria and are metabolically highly active, rendering them susceptible to mitochondrial dysfunction. Indeed, the causal role of mitochondrial dysfunction in SNHL progression is well established, and therefore, targeted for treatment. In this review, we aim to compile the emerging findings in the literature indicating the role of mitochondrial dysfunction in the progression of sensorineural hearing loss and highlight potential therapeutics targeting mitochondrial dysfunction for hearing loss treatment.


Subject(s)
Hearing Loss, Sensorineural , Neurodegenerative Diseases , Parkinson Disease , Humans , Mitochondria/metabolism , Hearing Loss, Sensorineural/metabolism , Neurodegenerative Diseases/metabolism , Reactive Oxygen Species/metabolism
4.
Nat Commun ; 12(1): 3091, 2021 05 25.
Article in English | MEDLINE | ID: mdl-34035268

ABSTRACT

VEGFR2 (KDR/Flk1) signaling in endothelial cells (ECs) plays a central role in angiogenesis. The P-type ATPase transporter ATP7A regulates copper homeostasis, and its role in VEGFR2 signaling and angiogenesis is entirely unknown. Here, we describe the unexpected crosstalk between the Copper transporter ATP7A, autophagy, and VEGFR2 degradation. The functional significance of this Copper transporter was demonstrated by the finding that inducible EC-specific ATP7A deficient mice or ATP7A-dysfunctional ATP7Amut mice showed impaired post-ischemic neovascularization. In ECs, loss of ATP7A inhibited VEGF-induced VEGFR2 signaling and angiogenic responses, in part by promoting ligand-induced VEGFR2 protein degradation. Mechanistically, VEGF stimulated ATP7A translocation from the trans-Golgi network to the plasma membrane where it bound to VEGFR2, which prevented autophagy-mediated lysosomal VEGFR2 degradation by inhibiting autophagic cargo/adapter p62/SQSTM1 binding to ubiquitinated VEGFR2. Enhanced autophagy flux due to ATP7A dysfunction in vivo was confirmed by autophagy reporter CAG-ATP7Amut -RFP-EGFP-LC3 transgenic mice. In summary, our study uncovers a novel function of ATP7A to limit autophagy-mediated degradation of VEGFR2, thereby promoting VEGFR2 signaling and angiogenesis, which restores perfusion recovery and neovascularization. Thus, endothelial ATP7A is identified as a potential therapeutic target for treatment of ischemic cardiovascular diseases.


Subject(s)
Autophagy/genetics , Blood Vessels/metabolism , Copper-Transporting ATPases/genetics , P-type ATPases/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Animals , Blood Vessels/drug effects , Blood Vessels/physiology , COS Cells , Cells, Cultured , Chlorocebus aethiops , Copper-Transporting ATPases/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/physiology , Humans , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , P-type ATPases/metabolism , RNA Interference , Signal Transduction/genetics , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism
5.
Nucleic Acids Res ; 49(5): 2418-2434, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33590097

ABSTRACT

Cockayne syndrome (CS) is a segmental premature aging syndrome caused primarily by defects in the CSA or CSB genes. In addition to premature aging, CS patients typically exhibit microcephaly, progressive mental and sensorial retardation and cutaneous photosensitivity. Defects in the CSB gene were initially thought to primarily impair transcription-coupled nucleotide excision repair (TC-NER), predicting a relatively consistent phenotype among CS patients. In contrast, the phenotypes of CS patients are pleiotropic and variable. The latter is consistent with recent work that implicates CSB in multiple cellular systems and pathways, including DNA base excision repair, interstrand cross-link repair, transcription, chromatin remodeling, RNAPII processing, nucleolin regulation, rDNA transcription, redox homeostasis, and mitochondrial function. The discovery of additional functions for CSB could potentially explain the many clinical phenotypes of CSB patients. This review focuses on the diverse roles played by CSB in cellular pathways that enhance genome stability, providing insight into the molecular features of this complex premature aging disease.


Subject(s)
DNA Helicases/physiology , DNA Repair Enzymes/physiology , Poly-ADP-Ribose Binding Proteins/physiology , Chromatin Assembly and Disassembly , DNA Breaks, Double-Stranded , DNA Helicases/chemistry , DNA Repair , DNA Repair Enzymes/chemistry , DNA, Ribosomal/biosynthesis , Gene Expression Regulation , Humans , Mitochondria/genetics , Mitochondria/metabolism , Poly-ADP-Ribose Binding Proteins/chemistry , RNA Polymerase II/metabolism , Transcription, Genetic
6.
Aging Cell ; 19(12): e13268, 2020 12.
Article in English | MEDLINE | ID: mdl-33166073

ABSTRACT

Cockayne syndrome (CS) is a rare premature aging disease, most commonly caused by mutations of the genes encoding the CSA or CSB proteins. CS patients display cachectic dwarfism and severe neurological manifestations and have an average life expectancy of 12 years. The CS proteins are involved in transcription and DNA repair, with the latter including transcription-coupled nucleotide excision repair (TC-NER). However, there is also evidence for mitochondrial dysfunction in CS, which likely contributes to the severe premature aging phenotype of this disease. While damaged mitochondria and impaired mitophagy were characterized in mice with CSB deficiency, such changes in the CS nematode model and CS patients are not fully known. Our cross-species transcriptomic analysis in CS postmortem brain tissue, CS mouse, and nematode models shows that mitochondrial dysfunction is indeed a common feature in CS. Restoration of mitochondrial dysfunction through NAD+ supplementation significantly improved lifespan and healthspan in the CS nematodes, highlighting mitochondrial dysfunction as a major driver of the aging features of CS. In cerebellar samples from CS patients, we found molecular signatures of dysfunctional mitochondrial dynamics and impaired mitophagy/autophagy. In primary cells depleted for CSA or CSB, this dysfunction can be corrected with supplementation of NAD+ precursors. Our study provides support for the interconnection between major causative aging theories, DNA damage accumulation, mitochondrial dysfunction, and compromised mitophagy/autophagy. Together, these three agents contribute to an accelerated aging program that can be averted by cellular NAD+ restoration.


Subject(s)
Cockayne Syndrome/metabolism , DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , Mitochondria/metabolism , NAD/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Transcription Factors/metabolism , AMP-Activated Protein Kinases/metabolism , Aging, Premature/genetics , Aging, Premature/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Cerebellum/metabolism , Cockayne Syndrome/genetics , Cockayne Syndrome/pathology , DNA Helicases/deficiency , DNA Helicases/genetics , DNA Repair Enzymes/deficiency , DNA Repair Enzymes/genetics , Disease Models, Animal , Humans , Longevity/genetics , Longevity/physiology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mitochondria/pathology , Oligonucleotide Array Sequence Analysis , Poly-ADP-Ribose Binding Proteins/deficiency , Poly-ADP-Ribose Binding Proteins/genetics , Signal Transduction , Transcription Factors/deficiency , Transcription Factors/genetics
7.
Am J Physiol Cell Physiol ; 319(5): C933-C944, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32936699

ABSTRACT

Caveolin-1 (Cav-1) is a scaffolding protein and a major component of caveolae/lipid rafts. Previous reports have shown that endothelial dysfunction in Cav-1-deficient (Cav-1-/-) mice is mediated by elevated oxidative stress through endothelial nitric oxide synthase (eNOS) uncoupling and increased NADPH oxidase. Oxidant stress is the net balance of oxidant generation and scavenging, and the role of Cav-1 as a regulator of antioxidant enzymes in vascular tissue is poorly understood. Extracellular SOD (SOD3) is a copper (Cu)-containing enzyme that is secreted from vascular smooth muscle cells/fibroblasts and subsequently binds to the endothelial cells surface, where it scavenges extracellular [Formula: see text] and preserves endothelial function. SOD3 activity is dependent on Cu, supplied by the Cu transporter ATP7A, but whether Cav-1 regulates the ATP7A-SOD3 axis and its role in oxidative stress-mediated vascular dysfunction has not been studied. Here we show that the activity of SOD3, but not SOD1, was significantly decreased in Cav-1-/- vessels, which was rescued by re-expression of Cav-1 or Cu supplementation. Loss of Cav-1 reduced ATP7A protein, but not mRNA, and this was mediated by ubiquitination of ATP7A and proteasomal degradation. ATP7A bound to Cav-1 and was colocalized with SOD3 in caveolae/lipid rafts or perinucleus in vascular tissues or cells. Impaired endothelium-dependent vasorelaxation in Cav-1-/- mice was rescued by gene transfer of SOD3 or by ATP7A-overexpressing transgenic mice. These data reveal an unexpected role of Cav-1 in stabilizing ATP7A protein expression by preventing its ubiquitination and proteasomal degradation, thereby increasing SOD3 activity, which in turn protects against vascular oxidative stress-mediated endothelial dysfunction.


Subject(s)
Caveolin 1/genetics , Copper-Transporting ATPases/genetics , Endothelial Cells/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase/genetics , Animals , Aorta/cytology , Aorta/metabolism , Caveolin 1/deficiency , Copper/pharmacology , Copper Transport Proteins/genetics , Copper Transport Proteins/metabolism , Copper-Transporting ATPases/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation , Male , Mesenteric Arteries/cytology , Mesenteric Arteries/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Oxidative Stress , Primary Cell Culture , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Signal Transduction , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/metabolism , Ubiquitination/drug effects , Vasodilation/drug effects
8.
Nucleic Acids Res ; 48(5): 2473-2485, 2020 03 18.
Article in English | MEDLINE | ID: mdl-31970402

ABSTRACT

Cockayne Syndrome (CS) is a rare neurodegenerative disease characterized by short stature, accelerated aging and short lifespan. Mutations in two human genes, ERCC8/CSA and ERCC6/CSB, are causative for CS and their protein products, CSA and CSB, while structurally unrelated, play roles in DNA repair and other aspects of DNA metabolism in human cells. Many clinical and molecular features of CS remain poorly understood, and it was observed that CSA and CSB regulate transcription of ribosomal DNA (rDNA) genes and ribosome biogenesis. Here, we investigate the dysregulation of rRNA synthesis in CS. We report that Nucleolin (Ncl), a nucleolar protein that regulates rRNA synthesis and ribosome biogenesis, interacts with CSA and CSB. In addition, CSA induces ubiquitination of Ncl, enhances binding of CSB to Ncl, and CSA and CSB both stimulate the binding of Ncl to rDNA and subsequent rRNA synthesis. CSB and CSA also increase RNA Polymerase I loading to the coding region of the rDNA and this is Ncl dependent. These findings suggest that CSA and CSB are positive regulators of rRNA synthesis via Ncl regulation. Most CS patients carry mutations in CSA and CSB and present with similar clinical features, thus our findings provide novel insights into disease mechanism.


Subject(s)
Cockayne Syndrome/genetics , DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , Gene Expression Regulation , Phosphoproteins/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , RNA, Ribosomal/genetics , RNA-Binding Proteins/genetics , Transcription Factors/metabolism , Transcription, Genetic , Cell Line , DNA, Ribosomal/genetics , Humans , Models, Biological , Phosphoproteins/metabolism , Protein Binding , RNA-Binding Proteins/metabolism , Nucleolin
9.
NPJ Aging Mech Dis ; 6: 1, 2020.
Article in English | MEDLINE | ID: mdl-31934345

ABSTRACT

Age-related hearing loss (ARHL) is one of the most common disorders affecting elderly individuals. There is an urgent need for effective preventive measures for ARHL because none are currently available. Cockayne syndrome (CS) is a premature aging disease that presents with progressive hearing loss at a young age, but is otherwise similar to ARHL. There are two human genetic complementation groups of CS, A and B. While the clinical phenotypes in patients are similar, the proteins have very diverse functions, and insight into their convergence is of great interest. Here, we use mouse models for CS (CSA -/- and CSB m/m ) that recapitulate the hearing loss in human CS patients. We previously showed that NAD+, a key metabolite with various essential functions, is reduced in CS and associated with multiple CS phenotypes. In this study, we report that NAD+ levels are reduced in the cochlea of CSB m/m mice and that short-term treatment (10 days) with the NAD+ precursor nicotinamide riboside (NR), prevents hearing loss, restores outer hair cell loss, and improves cochlear health in CSB m/m mice. Similar, but more modest effects were observed in CSA -/- mice. Remarkably, we observed a reduction in synaptic ribbon counts in the presynaptic zones of inner hair cells in both CSA -/- and CSB m/m mice, pointing to a converging mechanism for cochlear defects in CS. Ribbon synapses facilitate rapid and sustained synaptic transmission over long periods of time. Ribeye, a core protein of synaptic ribbons, possesses an NAD(H) binding pocket which regulates its activity. Intriguingly, NAD+ supplementation rescues reduced synaptic ribbon formation in both CSA -/- and CSB m/m mutant cochleae. These findings provide valuable insight into the mechanism of CS- and ARHL-associated hearing loss, and suggest a possible intervention.

10.
Nat Commun ; 10(1): 5284, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31754102

ABSTRACT

Metabolic dysfunction is a primary feature of Werner syndrome (WS), a human premature aging disease caused by mutations in the gene encoding the Werner (WRN) DNA helicase. WS patients exhibit severe metabolic phenotypes, but the underlying mechanisms are not understood, and whether the metabolic deficit can be targeted for therapeutic intervention has not been determined. Here we report impaired mitophagy and depletion of NAD+, a fundamental ubiquitous molecule, in WS patient samples and WS invertebrate models. WRN regulates transcription of a key NAD+ biosynthetic enzyme nicotinamide nucleotide adenylyltransferase 1 (NMNAT1). NAD+ repletion restores NAD+ metabolic profiles and improves mitochondrial quality through DCT-1 and ULK-1-dependent mitophagy. At the organismal level, NAD+ repletion remarkably extends lifespan and delays accelerated aging, including stem cell dysfunction, in Caenorhabditis elegans and Drosophila melanogaster models of WS. Our findings suggest that accelerated aging in WS is mediated by impaired mitochondrial function and mitophagy, and that bolstering cellular NAD+ levels counteracts WS phenotypes.


Subject(s)
Aging, Premature/metabolism , Mitophagy , NAD/metabolism , Werner Syndrome Helicase/metabolism , Werner Syndrome/metabolism , Aging, Premature/genetics , Animals , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Disease Models, Animal , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mutation , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Werner Syndrome/genetics , Werner Syndrome Helicase/genetics
11.
Nucleic Acids Res ; 47(16): 8548-8562, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31276581

ABSTRACT

Cockayne syndrome is an accelerated aging disorder, caused by mutations in the CSA or CSB genes. In CSB-deficient cells, poly (ADP ribose) polymerase (PARP) is persistently activated by unrepaired DNA damage and consumes and depletes cellular nicotinamide adenine dinucleotide, which leads to mitochondrial dysfunction. Here, the distribution of poly (ADP ribose) (PAR) was determined in CSB-deficient cells using ADPr-ChAP (ADP ribose-chromatin affinity purification), and the results show striking enrichment of PAR at transcription start sites, depletion of heterochromatin and downregulation of H3K9me3-specific methyltransferases SUV39H1 and SETDB1. Induced-expression of SETDB1 in CSB-deficient cells downregulated PAR and normalized mitochondrial function. The results suggest that defects in CSB are strongly associated with loss of heterochromatin, downregulation of SETDB1, increased PAR in highly-transcribed regions, and mitochondrial dysfunction.


Subject(s)
Cellular Senescence/genetics , Cockayne Syndrome/genetics , DNA Helicases/genetics , DNA Repair Enzymes/genetics , Histones/genetics , Mitochondria/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , Protein Methyltransferases/genetics , Transcription Factors/genetics , Cell Line, Transformed , Chromatin/chemistry , Chromatin/metabolism , Cockayne Syndrome/metabolism , Cockayne Syndrome/pathology , DNA/genetics , DNA/metabolism , DNA Damage , DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Histone-Lysine N-Methyltransferase , Histones/metabolism , Humans , Methyltransferases/genetics , Methyltransferases/metabolism , Mitochondria/pathology , Mutation , NAD/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Protein Methyltransferases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Transcription Initiation Site , Transcription, Genetic
12.
FEBS J ; 286(6): 1058-1073, 2019 03.
Article in English | MEDLINE | ID: mdl-30238623

ABSTRACT

The biology of aging is an area of intense research, and many questions remain about how and why cell and organismal functions decline over time. In mammalian cells, genomic instability and mitochondrial dysfunction are thought to be among the primary drivers of cellular aging. This review focuses on the interrelationship between genomic instability and mitochondrial dysfunction in mammalian cells and its relevance to age-related functional decline at the molecular and cellular level. The importance of oxidative stress and key DNA damage response pathways in cellular aging is discussed, with a special focus on poly (ADP-ribose) polymerase 1, whose persistent activation depletes cellular energy reserves, leading to mitochondrial dysfunction, loss of energy homeostasis, and altered cellular metabolism. Elucidation of the relationship between genomic instability, mitochondrial dysfunction, and the signaling pathways that connect these pathways/processes are keys to the future of research on human aging. An important component of mitochondrial health preservation is mitophagy, and this and other areas that are particularly ripe for future investigation will be discussed.


Subject(s)
Aging/pathology , Genomic Instability , Homeostasis , Mitochondria/pathology , Oxidative Stress , Aging/metabolism , Animals , Energy Metabolism , Humans , Mitochondria/metabolism , Mitophagy , Poly(ADP-ribose) Polymerases/metabolism
13.
Sci Rep ; 8(1): 17490, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30504782

ABSTRACT

Cockayne syndrome (CS) is an inherited disorder that involves photosensitivity, developmental defects, progressive degeneration and characteristics of premature aging. Evidence indicates primarily nuclear roles for the major CS proteins, CSA and CSB, specifically in DNA repair and RNA transcription. We reveal herein a complex regulation of CSB targeting that involves three major consensus signals: NLS1 (aa467-481), which directs nuclear and nucleolar localization in cooperation with NoLS1 (aa302-341), and NLS2 (aa1038-1055), which seemingly optimizes nuclear enrichment. CSB localization to the nucleolus was also found to be important for full UVC resistance. CSA, which does not contain any obvious targeting sequences, was adversely affected (i.e. presumably destabilized) by any form of truncation. No inter-coordination between the subnuclear localization of CSA and CSB was observed, implying that this aspect does not underlie the clinical features of CS. The E3 ubiquitin ligase binding partner of CSA, DDB1, played an important role in CSA stability (as well as DDB2), and facilitated CSA association with chromatin following UV irradiation; yet did not affect CSB chromatin binding. We also observed that initial recruitment of CSB to DNA interstrand crosslinks is similar in the nucleoplasm and nucleolus, although final accumulation is greater in the former. Whereas assembly of CSB at sites of DNA damage in the nucleolus was not affected by RNA polymerase I inhibition, stable retention at these sites of presumed repair was abrogated. Our studies reveal a multi-faceted regulation of the intranuclear dynamics of CSA and CSB that plays a role in mediating their cellular functions.


Subject(s)
Biomarkers , Cell Nucleus/metabolism , Cockayne Syndrome/metabolism , Amino Acid Sequence , Cockayne Syndrome/etiology , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Fluorescent Antibody Technique , Genes, Reporter , Humans , Intracellular Space , Mutation , Protein Sorting Signals , Protein Transport , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Arterioscler Thromb Vasc Biol ; 38(3): 529-541, 2018 03.
Article in English | MEDLINE | ID: mdl-29301787

ABSTRACT

OBJECTIVE: Copper transporter ATP7A (copper-transporting/ATPase) is required for full activation of SOD3 (extracellular superoxide dismutase), which is secreted from vascular smooth muscle cells (VSMCs) and anchors to endothelial cell surface to preserve endothelial function by scavenging extracellular superoxide. We reported that ATP7A protein expression and SOD3 activity are decreased in insulin-deficient type 1 diabetes mellitus vessels, thereby, inducing superoxide-mediated endothelial dysfunction, which are rescued by insulin treatment. However, it is unknown regarding the mechanism by which insulin increases ATP7A expression in VSMCs and whether ATP7A downregulation is observed in T2DM (type2 diabetes mellitus) mice and human in which insulin-Akt (protein kinase B) pathway is selectively impaired. APPROACH AND RESULTS: Here we show that ATP7A protein is markedly downregulated in vessels isolated from T2DM patients, as well as those from high-fat diet-induced or db/db T2DM mice. Akt2 (protein kinase B beta) activated by insulin promotes ATP7A stabilization via preventing ubiquitination/degradation as well as translocation to plasma membrane in VSMCs, which contributes to activation of SOD3 that protects against T2DM-induced endothelial dysfunction. Downregulation of ATP7A in T2DM vessels is restored by constitutive active Akt or PTP1B-/- (protein-tyrosine phosphatase 1B-deficient) T2DM mice, which enhance insulin-Akt signaling. Immunoprecipitation, in vitro kinase assay, and mass spectrometry analysis reveal that insulin stimulates Akt2 binding to ATP7A to induce phosphorylation at Ser1424/1463/1466. Furthermore, SOD3 activity is reduced in Akt2-/- vessels or VSMCs, which is rescued by ATP7A overexpression. CONCLUSION: Akt2 plays a critical role in ATP7A protein stabilization and translocation to plasma membrane in VSMCs, which contributes to full activation of vascular SOD3 that protects against endothelial dysfunction in T2DM.


Subject(s)
Copper-Transporting ATPases/metabolism , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Type 2/enzymology , Diabetic Angiopathies/enzymology , Endothelium, Vascular/enzymology , Muscle, Smooth, Vascular/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Superoxide Dismutase/metabolism , Animals , Aorta, Thoracic/enzymology , Aorta, Thoracic/physiopathology , Cells, Cultured , Copper-Transporting ATPases/genetics , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Diabetic Angiopathies/genetics , Diabetic Angiopathies/physiopathology , Diabetic Angiopathies/prevention & control , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Enzyme Stability , Female , Humans , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Male , Mesenteric Arteries/enzymology , Mesenteric Arteries/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Phosphorylation , Protein Transport , Proto-Oncogene Proteins c-akt/deficiency , Proto-Oncogene Proteins c-akt/genetics , Rats, Sprague-Dawley , Signal Transduction , Superoxide Dismutase/deficiency , Superoxide Dismutase/genetics , Vasodilation
15.
Nat Commun ; 8(1): 2039, 2017 12 11.
Article in English | MEDLINE | ID: mdl-29229926

ABSTRACT

Pathway choice within DNA double-strand break (DSB) repair is a tightly regulated process to maintain genome integrity. RECQL4, deficient in Rothmund-Thomson Syndrome, promotes the two major DSB repair pathways, non-homologous end joining (NHEJ) and homologous recombination (HR). Here we report that RECQL4 promotes and coordinates NHEJ and HR in different cell cycle phases. RECQL4 interacts with Ku70 to promote NHEJ in G1 when overall cyclin-dependent kinase (CDK) activity is low. During S/G2 phases, CDK1 and CDK2 (CDK1/2) phosphorylate RECQL4 on serines 89 and 251, enhancing MRE11/RECQL4 interaction and RECQL4 recruitment to DSBs. After phosphorylation, RECQL4 is ubiquitinated by the DDB1-CUL4A E3 ubiquitin ligase, which facilitates its accumulation at DSBs. Phosphorylation of RECQL4 stimulates its helicase activity, promotes DNA end resection, increases HR and cell survival after ionizing radiation, and prevents cellular senescence. Collectively, we propose that RECQL4 modulates the pathway choice of NHEJ and HR in a cell cycle-dependent manner.


Subject(s)
Cell Cycle , DNA Breaks, Double-Stranded , DNA End-Joining Repair , RecQ Helicases/metabolism , Recombinational DNA Repair , Ubiquitination , Cell Line, Tumor , Cullin Proteins/genetics , Cullin Proteins/metabolism , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , HEK293 Cells , Humans , Ku Autoantigen/genetics , Ku Autoantigen/metabolism , Phosphorylation , Protein Binding , RNA Interference , RecQ Helicases/genetics
16.
Nat Chem Biol ; 13(1): 62-68, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27820802

ABSTRACT

RAS GTPases are important mediators of oncogenesis in humans. However, pharmacological inhibition of RAS has proved challenging. Here we describe a functionally critical region, located outside the effector lobe of RAS, that can be targeted for inhibition. We developed NS1, a synthetic binding protein (monobody) that bound with high affinity to both GTP- and GDP-bound states of H-RAS and K-RAS but not N-RAS. NS1 potently inhibited growth factor signaling and oncogenic H-RAS- and K-RAS-mediated signaling and transformation but did not block oncogenic N-RAS, BRAF or MEK1. NS1 bound the α4-ß6-α5 region of RAS, which disrupted RAS dimerization and nanoclustering and led to blocking of CRAF-BRAF heterodimerization and activation. These results establish the importance of the α4-ß6-α5 interface in RAS-mediated signaling and define a previously unrecognized site in RAS for inhibiting RAS function.


Subject(s)
Allosteric Site/drug effects , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , ras Proteins/antagonists & inhibitors , ras Proteins/chemistry , Animals , Antibodies, Monoclonal/chemistry , COS Cells , Cells, Cultured , Chlorocebus aethiops , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , ras Proteins/metabolism
17.
Cancer Lett ; 359(2): 262-8, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25622909

ABSTRACT

Phosphatidylinositol 3-kinases (PI3Ks) play important roles in human tumorigenesis. Activation of the PI3K target AKT is frequent in neuroblastoma (NB) and correlates with poor prognosis. PI3K pan-inhibitors reduce NB tumor formation but present severe toxicity, which limits their therapeutic potential. Therefore, defining the importance of specific PI3K isoforms may aid in developing more effective therapeutic strategies. We previously demonstrated that PI3K Class IIß (PI3KC2ß) and its regulator intersectin 1 (ITSN1) are highly expressed in primary NB tumors and cell lines. Silencing ITSN1 dramatically reduced the tumorigenic potential of NB cells. Interestingly, overexpression of PI3KC2ß rescued the anchorage-independent growth of ITSN1-silenced cells suggesting that PI3KC2ß mediates ITSN1's function in NB cells. To address the importance of PI3KC2ß in NBs, we generated PI3KC2ß-silenced lines and examined their biologic activity. Herein, we demonstrate that PI3KC2ß-silencing inhibits early stages of NB tumorigenic growth. We also show that loss of endogenous PI3KC2ß or ITSN1 reduces AKT activation but does not impact ERK-MAPK activation. These data reveal a novel role for PI3KC2ß in human NB tumorigenesis.


Subject(s)
Carcinogenesis/metabolism , Neuroblastoma/enzymology , Phosphatidylinositol 3-Kinases/physiology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Class II Phosphatidylinositol 3-Kinases , Gene Knockdown Techniques , Humans , Isoenzymes/physiology , MAP Kinase Signaling System , Mice, Nude , Neoplasm Transplantation , Neuroblastoma/pathology , Proto-Oncogene Proteins c-akt/metabolism
18.
Mol Cell Biol ; 34(2): 271-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24216759

ABSTRACT

Ubiquitylation of receptor tyrosine kinases (RTKs) regulates their trafficking and lysosomal degradation. The multidomain scaffolding protein intersectin 1 (ITSN1) is an important regulator of this process. ITSN1 stimulates ubiquitylation of the epidermal growth factor receptor (EGFR) through enhancing the activity of the Cbl E3 ubiquitin ligase. However, the precise mechanism through which ITSN1 enhances Cbl activity is unclear. Here, we demonstrate that ITSN1 interacts with and recruits the Shp2 tyrosine phosphatase to Spry2 to enhance its dephosphorylation, thereby disrupting the inhibitory effect of Spry2 on Cbl and enhancing EGFR ubiquitylation. In contrast, expression of a catalytically inactive Shp2 mutant reversed the effect of ITSN1 on Spry2 dephosphorylation and decreased Cbl-mediated EGFR ubiquitylation. In addition, disruption of ITSN1 binding to Spry2 through point mutation of the Pro-rich ITSN1 binding site in Spry2 resulted in decreased Shp2-Spry2 interaction and enhanced Spry2 tyrosine phosphorylation. This study demonstrates that ITSN1 enhances Cbl activity, in part, by modulating the interaction of Cbl with Spry2 through recruitment of Shp2 phosphatase to the Cbl-Spry2 complex. These findings reveal a new level of complexity in the regulation of RTKs by Cbl through ITSN1 binding with Shp2 and Spry2.


Subject(s)
Adaptor Proteins, Vesicular Transport/physiology , ErbB Receptors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Ubiquitination , Animals , COS Cells , Chlorocebus aethiops , Epidermal Growth Factor/physiology , Humans , Mice , Mice, Knockout , Phosphorylation , Protein Binding , Protein Isoforms , Protein Transport , Receptor Protein-Tyrosine Kinases/metabolism
19.
PLoS One ; 7(4): e36023, 2012.
Article in English | MEDLINE | ID: mdl-22558309

ABSTRACT

Members of the intersectin (ITSN) family of scaffold proteins consist of multiple modular domains, each with distinct ligand preferences. Although ITSNs were initially implicated in the regulation of endocytosis, subsequent studies have revealed a more complex role for these scaffold proteins in regulation of additional biochemical pathways. In this study, we performed a high throughput yeast two-hybrid screen to identify additional pathways regulated by these scaffolds. Although several known ITSN binding partners were identified, we isolated more than 100 new targets for the two mammalian ITSN proteins, ITSN1 and ITSN2. We present the characterization of several of these new targets which implicate ITSNs in the regulation of the Rab and Arf GTPase pathways as well as regulation of the disrupted in schizophrenia 1 (DISC1) interactome. In addition, we demonstrate that ITSN proteins form homomeric and heteromeric complexes with each other revealing an added level of complexity in the function of these evolutionarily conserved scaffolds.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Multigene Family , Protein Interaction Maps , Animals , COS Cells , Chlorocebus aethiops , Endocytosis , GTP Phosphohydrolases/metabolism , HEK293 Cells , High-Throughput Screening Assays , Humans , Nervous System Diseases/metabolism , Phosphatidylinositols/metabolism , Protein Binding , Protein Multimerization , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Two-Hybrid System Techniques
20.
Mol Cell Biol ; 32(4): 817-25, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22158968

ABSTRACT

Ubiquitylation of receptor tyrosine kinases plays a critical role in regulating the trafficking and lysosomal degradation of these important signaling molecules. We identified the multidomain scaffolding protein intersectin 1 (ITSN1) as an important regulator of this process (N. P. Martin et al., Mol. Pharmacol. 70:1463-1653, 2006) ITSN1 stimulates ubiquitylation of the epidermal growth factor receptor (EGFR) through enhancing the activity of the Cbl E3 ubiquitin ligase. However, the precise mechanism through which ITSN1 enhances Cbl activity was unclear. In this study, we found that ITSN1 enhances Cbl activity through disrupting the interaction of Cbl with the Sprouty2 (Spry2) inhibitory protein. We demonstrate that ITSN1 binds Pro-rich regions in both Cbl and Spry2 and that interaction of ITSN1 with Spry2 disrupts Spry2-Cbl interaction, resulting in enhanced ubiquitylation of the EGFR. Disruption of ITSN1 binding to Spry2 through point mutation of the Pro-rich ITSN1 binding site in Spry2 results in enhanced Cbl-Spry2 interaction and inhibition of receptor ubiquitylation. This study demonstrates that ITSN1 enhances Cbl activity by modulating the interaction of Cbl with Spry2. In addition, our results reveal a new level of complexity in the regulation of Cbl through the interaction with ITSN1 and Spry2.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , ErbB Receptors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/genetics , Amino Acid Substitution , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , DNA Primers/genetics , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mutagenesis, Site-Directed , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-cbl/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Two-Hybrid System Techniques , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...