144 results on '"Croteau DL"'
Search Results
2. Loss of DNA glycosylases improves health and cognitive function in a C. elegans model of human tauopathy.
- Author
-
Tiwari V, Buvarp E, Borbolis F, Puligilla C, Croteau DL, Palikaras K, and Bohr VA
- Subjects
- Animals, Humans, Cognition, Mitochondria genetics, Mitochondria metabolism, Alzheimer Disease genetics, Alzheimer Disease metabolism, Alzheimer Disease pathology, Longevity genetics, Caenorhabditis elegans genetics, Disease Models, Animal, Tauopathies genetics, Tauopathies metabolism, Caenorhabditis elegans Proteins metabolism, Caenorhabditis elegans Proteins genetics, tau Proteins metabolism, tau Proteins genetics, DNA Glycosylases metabolism, DNA Glycosylases genetics, Animals, Genetically Modified
- Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder representing a major burden on families and society. Some of the main pathological hallmarks of AD are the accumulation of amyloid plaques (Aβ) and tau neurofibrillary tangles. However, it is still unclear how Aβ and tau aggregates promote specific phenotypic outcomes and lead to excessive oxidative DNA damage, neuronal cell death and eventually to loss of memory. Here we utilized a Caenorhabditis elegans (C. elegans) model of human tauopathy to investigate the role of DNA glycosylases in disease development and progression. Transgenic nematodes expressing a pro-aggregate form of tau displayed altered mitochondrial content, decreased lifespan, and cognitive dysfunction. Genetic ablation of either of the two DNA glycosylases found in C. elegans, NTH-1 and UNG-1, improved mitochondrial function, lifespan, and memory impairment. NTH-1 depletion resulted in a dramatic increase of differentially expressed genes, which was not apparent in UNG-1 deficient nematodes. Our findings clearly show that in addition to its enzymatic activity, NTH-1 has non-canonical functions highlighting its modulation as a potential therapeutic intervention to tackle tau-mediated pathology., (Published by Oxford University Press on behalf of Nucleic Acids Research 2024.)
- Published
- 2024
- Full Text
- View/download PDF
3. Nicotinamide riboside modulates the reactive species interactome, bioenergetic status and proteomic landscape in a brain-region-specific manner.
- Author
-
Marmolejo-Garza A, Chatre L, Croteau DL, Herron-Bedoya A, Luu MDA, Bernay B, Pontin J, Bohr VA, Boddeke E, and Dolga AM
- Subjects
- Animals, Mice, Proteomics, Proteome metabolism, Proteome drug effects, Oxidative Stress drug effects, Oxidative Stress physiology, Mice, Inbred C57BL, Male, Reactive Oxygen Species metabolism, Hippocampus metabolism, Hippocampus drug effects, Neurons metabolism, Neurons drug effects, Niacinamide analogs & derivatives, Niacinamide pharmacology, Pyridinium Compounds, Brain metabolism, Brain drug effects, Alzheimer Disease metabolism, Energy Metabolism drug effects, Energy Metabolism physiology
- Abstract
Nicotinamide riboside (NR), a precursor of nicotinamide adenine dinucleotide (NAD+), has robust cognitive benefits and alleviates neuroinflammation in Alzheimer's Disease (AD) mouse models without decreasing beta-amyloid plaque pathology. Such effects may be mediated by the reactive species interactome (RSI), at the metabolome level. In this study, we employed in vitro and in vivo models of oxidative stress, aging and AD to profile the effects of NR on neuronal survival, RSI, and the whole proteome characterization of cortex and hippocampus. RSI analysis yielded a complex modulation upon NR treatment. We constructed protein co-expression networks and correlated them to NR treatment and all measured reactive species. We observed brain-area specific effects of NR on co-expressed protein modules of oxidative phosphorylation, fatty acid oxidation, and neurotransmitter regulation pathways, which correlated with RSI components. The current study contributes to the understanding of modulation of the metabolome, specifically after NR treatment in AD and how it may play disease-modifying roles., Competing Interests: Declaration of competing interest V.A.B. has CRADA arrangements with ChromaDex. All other authors declare no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
4. Urolithin A improves Alzheimer's disease cognition and restores mitophagy and lysosomal functions.
- Author
-
Hou Y, Chu X, Park JH, Zhu Q, Hussain M, Li Z, Madsen HB, Yang B, Wei Y, Wang Y, Fang EF, Croteau DL, and Bohr VA
- Subjects
- Animals, Mice, Amyloid beta-Peptides metabolism, Cognition drug effects, Alzheimer Disease drug therapy, Coumarins pharmacology, Coumarins therapeutic use, Lysosomes drug effects, Lysosomes metabolism, Mice, Transgenic, Disease Models, Animal, Mitophagy drug effects
- Abstract
Background: Compromised autophagy, including impaired mitophagy and lysosomal function, plays pivotal roles in Alzheimer's disease (AD). Urolithin A (UA) is a gut microbial metabolite of ellagic acid that stimulates mitophagy. The effects of UA's long-term treatment of AD and mechanisms of action are unknown., Methods: We addressed these questions in three mouse models of AD with behavioral, electrophysiological, biochemical, and bioinformatic approaches., Results: Long-term UA treatment significantly improved learning, memory, and olfactory function in different AD transgenic mice. UA also reduced amyloid beta (Aβ) and tau pathologies and enhanced long-term potentiation. UA induced mitophagy via increasing lysosomal functions. UA improved cellular lysosomal function and normalized lysosomal cathepsins, primarily cathepsin Z, to restore lysosomal function in AD, indicating the critical role of cathepsins in UA-induced therapeutic effects on AD., Conclusions: Our study highlights the importance of lysosomal dysfunction in AD etiology and points to the high translational potential of UA., Highlights: Long-term urolithin A (UA) treatment improved learning, memory, and olfactory function in Alzheimer's disease (AD) mice. UA restored lysosomal functions in part by regulating cathepsin Z (Ctsz) protein. UA modulates immune responses and AD-specific pathophysiological pathways., (© 2024 The Authors. Alzheimer's & Dementia published by Wiley Periodicals LLC on behalf of Alzheimer's Association.)
- Published
- 2024
- Full Text
- View/download PDF
5. The cGAS-STING signaling pathway is modulated by urolithin A.
- Author
-
Madsen HB, Park JH, Chu X, Hou Y, Li Z, Rasmussen LJ, Croteau DL, Bohr VA, and Akbari M
- Subjects
- Animals, Humans, DNA metabolism, Signal Transduction physiology, Immunity, Innate, Nucleotidyltransferases genetics, Inflammation, Coumarins
- Abstract
During aging, general cellular processes, including autophagic clearance and immunological responses become compromised; therefore, identifying compounds that target these cellular processes is an important approach to improve our health span. The innate immune cGAS-STING pathway has emerged as an important signaling system in the organismal defense against viral and bacterial infections, inflammatory responses to cellular damage, regulation of autophagy, and tumor immunosurveillance. These key functions of the cGAS-STING pathway make it an attractive target for pharmacological intervention in disease treatments and in controlling inflammation and immunity. Here, we show that urolithin A (UA), an ellagic acid metabolite, exerts a profound effect on the expression of STING and enhances cGAS-STING activation and cytosolic DNA clearance in human cell lines. Animal laboratory models and limited human trials have reported no obvious adverse effects of UA administration. Thus, the use of UA alone or in combination with other pharmacological compounds may present a potential therapeutic approach in the treatment of human diseases that involves aberrant activation of the cGAS-STING pathway or accumulation of cytosolic DNA and this warrants further investigation in relevant transgenic animal models., Competing Interests: Declaration of Competing Interest The authors declare no conflicts of interest., (Copyright © 2023 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
6. Early-Onset Hearing Loss in Mouse Models of Alzheimer's Disease and Increased DNA Damage in the Cochlea.
- Author
-
Park JH, Sahbaz BD, Pekhale K, Chu X, Okur MN, Grati M, Isgrig K, Chien W, Chrysostomou E, Sullivan L, Croteau DL, Manor U, and Bohr VA
- 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/Polβ
+/- . The 3xTgAD/Polβ+/- 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/Polβ+/- 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., Competing Interests: Conflict of Interest Statement V.A.B. had a material CRADA agreement with ChromaDex, Inc.- Published
- 2024
- Full Text
- View/download PDF
7. The RNA-binding motif protein 14 regulates telomere integrity at the interface of TERRA and telomeric R-loops.
- Author
-
Wang Y, Zhu W, Jang Y, Sommers JA, Yi G, Puligilla C, Croteau DL, Yang Y, Kai M, and Liu Y
- Subjects
- Humans, DNA genetics, R-Loop Structures, RNA genetics, RNA metabolism, RNA, Long Noncoding genetics, RNA-Binding Motifs, Neoplasms genetics, Telomere genetics, Telomere metabolism, Telomere Homeostasis
- Abstract
Telomeric repeat-containing RNA (TERRA) and its formation of RNA:DNA hybrids (or TERRA R-loops), influence telomere maintenance, particularly in human cancer cells that use homologous recombination-mediated alternative lengthening of telomeres. Here, we report that the RNA-binding motif protein 14 (RBM14) is associated with telomeres in human cancer cells. RBM14 negatively regulates TERRA expression. It also binds to TERRA and inhibits it from forming TERRA R-loops at telomeres. RBM14 depletion has several effects, including elevated TERRA levels, telomeric R-loops, telomere dysfunction-induced DNA damage foci formation, particularly in the presence of DNA replication stress, pRPA32 accumulation at telomeres and telomere signal-free ends. Thus, RBM14 protects telomere integrity via modulating TERRA levels and its R-loop formation at telomeres., (© The Author(s) 2023. Published by Oxford University Press on behalf of Nucleic Acids Research 2023.)
- Published
- 2023
- Full Text
- View/download PDF
8. Long-term NAD+ supplementation prevents the progression of age-related hearing loss in mice.
- Author
-
Okur MN, Sahbaz BD, Kimura R, Manor U, Patel J, Park JH, Andrade L, Puligilla C, Croteau DL, and Bohr VA
- Subjects
- Humans, Animals, Mice, Peroxisome Proliferator-Activated Receptors, Cochlea, Dietary Supplements, NAD, Presbycusis drug therapy, Presbycusis prevention & control
- 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., (© 2023 The Authors. Aging Cell published by Anatomical Society and John Wiley & Sons Ltd. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.)
- Published
- 2023
- Full Text
- View/download PDF
9. Mitochondrial OGG1 expression reduces age-associated neuroinflammation by regulating cytosolic mitochondrial DNA.
- Author
-
Hussain M, Chu X, Duan Sahbaz B, Gray S, Pekhale K, Park JH, Croteau DL, and Bohr VA
- Subjects
- Animals, Female, Male, Mice, DNA Damage, DNA Repair, Inflammation genetics, Inflammation metabolism, Mice, Transgenic, Mitochondria genetics, Mitochondria metabolism, Neuroinflammatory Diseases, Oxidative Stress genetics, Humans, DNA Glycosylases genetics, DNA Glycosylases metabolism, DNA, Mitochondrial genetics, DNA, Mitochondrial metabolism
- Abstract
Aging is accompanied by a decline in DNA repair efficiency, which leads to the accumulation of different types of DNA damage. Age-associated chronic inflammation and generation of reactive oxygen species exacerbate the aging process and age-related chronic disorders. These inflammatory processes establish conditions that favor accumulation of DNA base damage, especially 8-oxo-7,8 di-hydroguanine (8-oxoG), which in turn contributes to various age associated diseases. 8-oxoG is repaired by 8-oxoG glycosylase1 (OGG1) through the base excision repair (BER) pathway. OGG1 is present in both the cell nucleus and in mitochondria. Mitochondrial OGG1 has been implicated in mitochondrial DNA repair and increased mitochondrial function. Using transgenic mouse models and cell lines that have been engineered to have enhanced expression of mitochondria-targeted OGG1 (mtOGG1), we show that elevated levels of mtOGG1 in mitochondria can reverse aging-associated inflammation and improve functions. Old male mtOGG1
Tg mice show decreased inflammation response, decreased TNFα levels and multiple pro-inflammatory cytokines. Moreover, we observe that male mtOGG1Tg mice show resistance to STING activation. Interestingly, female mtOGG1Tg mice did not respond to mtOGG1 overexpression. Further, HMC3 cells expressing mtOGG1 display decreased release of mtDNA into the cytoplasm after lipopolysacchride induction and regulate inflammation through the pSTING pathway. Also, increased mtOGG1 expression reduced LPS-induced loss of mitochondrial functions. These results suggest that mtOGG1 regulates age-associated inflammation by controlling release of mtDNA into the cytoplasm., (Published by Elsevier Inc.)- Published
- 2023
- Full Text
- View/download PDF
10. RecQ dysfunction contributes to social and depressive-like behavior and affects aldolase activity in mice.
- Author
-
Hou Y, Park JH, Dan X, Chu X, Yang B, Hussain M, Croteau DL, and Bohr VA
- Subjects
- Animals, Humans, Mice, DNA Repair, DNA Damage, Genomic Instability, Aldehyde-Lyases genetics, Aldehyde-Lyases metabolism, RecQ Helicases genetics, RecQ Helicases metabolism, DNA Replication
- Abstract
RecQ helicase family proteins play vital roles in maintaining genome stability, including DNA replication, recombination, and DNA repair. In human cells, there are five RecQ helicases: RECQL1, Bloom syndrome (BLM), Werner syndrome (WRN), RECQL4, and RECQL5. Dysfunction or absence of RecQ proteins is associated with genetic disorders, tumorigenesis, premature aging, and neurodegeneration. The biochemical and biological roles of RecQ helicases are rather well established, however, there is no systematic study comparing the behavioral changes among various RecQ-deficient mice including consequences of exposure to DNA damage. Here, we investigated the effects of ionizing irradiation (IR) on three RecQ-deficient mouse models (RecQ1, WRN and RecQ4). We find abnormal cognitive behavior in RecQ-deficient mice in the absence of IR. Interestingly, RecQ dysfunction impairs social ability and induces depressive-like behavior in mice after a single exposure to IR, suggesting that RecQ proteins play roles in mood and cognition behavior. Further, transcriptomic and metabolomic analyses revealed significant alterations in RecQ-deficient mice, especially after IR exposure. In particular, pathways related to neuronal and microglial functions, DNA damage repair, cell cycle, and reactive oxygen responses were downregulated in the RecQ4 and WRN mice. In addition, increased DNA damage responses were found in RecQ-deficient mice. Notably, two genes, Aldolase Fructose-Bisphosphate B (Aldob) and NADPH Oxidase 4 (Nox4), were differentially expressed in RecQ-deficient mice. Our findings suggest that RecQ dysfunction contributes to social and depressive-like behaviors in mice, and that aldolase activity may be associated with these changes, representing a potential therapeutic target., Competing Interests: Declaration of Competing Interest All authors declare no conflicts of interest., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
11. Loss of smelling is an early marker of aging and is associated with inflammation and DNA damage in C57BL/6J mice.
- Author
-
Dan X, Yang B, McDevitt RA, Gray S, Chu X, Claybourne Q, Figueroa DM, Zhang Y, Croteau DL, and Bohr VA
- Subjects
- Humans, Mice, Animals, Mice, Inbred C57BL, NAD metabolism, Aging pathology, DNA Damage, Olfactory Bulb metabolism, Olfactory Bulb pathology, Inflammation metabolism, Smell, Olfaction Disorders diagnosis, Olfaction Disorders pathology
- Abstract
Olfactory dysfunction is a prevalent symptom and an early marker of age-related neurodegenerative diseases in humans, including Alzheimer's and Parkinson's Diseases. However, as olfactory dysfunction is also a common symptom of normal aging, it is important to identify associated behavioral and mechanistic changes that underlie olfactory dysfunction in nonpathological aging. In the present study, we systematically investigated age-related behavioral changes in four specific domains of olfaction and the molecular basis in C57BL/6J mice. Our results showed that selective loss of odor discrimination was the earliest smelling behavioral change with aging, followed by a decline in odor sensitivity and detection while odor habituation remained in old mice. Compared to behavioral changes related with cognitive and motor functions, smelling loss was among the earliest biomarkers of aging. During aging, metabolites related with oxidative stress, osmolytes, and infection became dysregulated in the olfactory bulb, and G protein coupled receptor-related signaling was significantly down regulated in olfactory bulbs of aged mice. Poly ADP-ribosylation levels, protein expression of DNA damage markers, and inflammation increased significantly in the olfactory bulb of older mice. Lower NAD
+ levels were also detected. Supplementation of NAD+ through NR in water improved longevity and partially enhanced olfaction in aged mice. Our studies provide mechanistic and biological insights into the olfaction decline during aging and highlight the role of NAD+ for preserving smelling function and general health., (Published 2023. This article is a U.S. Government work and is in the public domain in the USA. Aging Cell published by Anatomical Society and John Wiley & Sons Ltd.)- Published
- 2023
- Full Text
- View/download PDF
12. Mitochondrial PARP1 regulates NAD + -dependent poly ADP-ribosylation of mitochondrial nucleoids.
- Author
-
Lee JH, Hussain M, Kim EW, Cheng SJ, Leung AKL, Fakouri NB, Croteau DL, and Bohr VA
- Subjects
- Poly(ADP-ribose) Polymerase Inhibitors, Mitochondria metabolism, DNA, Mitochondrial genetics, DNA, Mitochondrial metabolism, Poly ADP Ribosylation, NAD metabolism
- Abstract
PARPs play fundamental roles in multiple DNA damage recognition and repair pathways. Persistent nuclear PARP activation causes cellular NAD
+ depletion and exacerbates cellular aging. However, very little is known about mitochondrial PARP (mtPARP) and poly ADP-ribosylation (PARylation). The existence of mtPARP is controversial, and the biological roles of mtPARP-induced mitochondrial PARylation are unclear. Here, we demonstrate the presence of PARP1 and PARylation in purified mitochondria. The addition of the PARP1 substrate NAD+ to isolated mitochondria induced PARylation, which was suppressed by treatment with the inhibitor olaparib. Mitochondrial PARylation was also evaluated by enzymatic labeling of terminal ADP-ribose (ELTA). To further confirm the presence of mtPARP1, we evaluated mitochondrial nucleoid PARylation by ADP ribose-chromatin affinity purification (ADPr-ChAP) and PARP1 chromatin immunoprecipitation (ChIP). We observed that NAD+ stimulated PARylation and TFAM occupancy on the mtDNA regulatory region D-loop, inducing mtDNA transcription. These findings suggest that PARP1 is integrally involved in mitochondrial PARylation and that NAD+ -dependent mtPARP1 activity contributes to mtDNA transcriptional regulation., (© 2022. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)- Published
- 2022
- Full Text
- View/download PDF
13. Nicotinamide adenine dinucleotide supplementation drives gut microbiota variation in Alzheimer's mouse model.
- Author
-
Chu X, Hou Y, Meng Q, Croteau DL, Wei Y, De S, Becker KG, and Bohr VA
- Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease. Growing evidence suggests an important role for gut dysbiosis and gut microbiota-host interactions in aging and neurodegeneration. Our previous works have demonstrated that supplementation with the nicotinamide adenine dinucleotide (NAD
+ ) precursor, nicotinamide riboside (NR), reduced the brain features of AD, including neuroinflammation, deoxyribonucleic acid (DNA) damage, synaptic dysfunction, and cognitive impairment. However, the impact of NR administration on the intestinal microbiota of AD remains unknown. In this study, we investigated the relationship between gut microbiota and NR treatment in APP/PS1 transgenic (AD) mice. Compared with wild type (WT) mice, the gut microbiota diversity in AD mice was lower and the microbiota composition and enterotype were significantly different. Moreover, there were gender differences in gut microbiome between female and male AD mice. After supplementation with NR for 8 weeks, the decreased diversity and perturbated microbial compositions were normalized in AD mice. This included the species Oscillospira , Butyricicoccus , Desulfovibrio , Bifidobacterium , Olsenella , Adlercreutzia , Bacteroides , Akkermansia , and Lactobacillus . Our results indicate an interplay between NR and host-microbiota in APP/PS1 mice, suggesting that the effect of NR on gut dysbiosis may be an important component in its therapeutic functions in AD., Competing Interests: Author VB had a CRADA agreement with Chromadex, Inc. but receives no personal financial benefit. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Chu, Hou, Meng, Croteau, Wei, De, Becker and Bohr.)- Published
- 2022
- Full Text
- View/download PDF
14. WRNing for the right DNA repair pathway choice.
- Author
-
Lee JH, Croteau DL, and Bohr VA
- Subjects
- RecQ Helicases genetics, RecQ Helicases metabolism, DNA Breaks, Double-Stranded, DNA Repair
- Published
- 2022
- Full Text
- View/download PDF
15. A brain proteomic signature of incipient Alzheimer's disease in young APOE ε4 carriers identifies novel drug targets.
- Author
-
Roberts JA, Varma VR, An Y, Varma S, Candia J, Fantoni G, Tiwari V, Anerillas C, Williamson A, Saito A, Loeffler T, Schilcher I, Moaddel R, Khadeer M, Lovett J, Tanaka T, Pletnikova O, Troncoso JC, Bennett DA, Albert MS, Yu K, Niu M, Haroutunian V, Zhang B, Peng J, Croteau DL, Resnick SM, Gorospe M, Bohr VA, Ferrucci L, and Thambisetty M
- Abstract
Aptamer-based proteomics revealed differentially abundant proteins in Alzheimer’s disease (AD) brains in the Baltimore Longitudinal Study of Aging and Religious Orders Study (mean age, 89 ± 9 years). A subset of these proteins was also differentially abundant in the brains of young APOE ε4 carriers relative to noncarriers (mean age, 39 ± 6 years). Several of these proteins represent targets of approved and experimental drugs for other indications and were validated using orthogonal methods in independent human brain tissue samples as well as in transgenic AD models. Using cell culture–based phenotypic assays, we showed that drugs targeting the cytokine transducer STAT3 and the Src family tyrosine kinases, YES1 and FYN, rescued molecular phenotypes relevant to AD pathogenesis. Our findings may accelerate the development of effective interventions targeting the earliest molecular triggers of AD.
- Published
- 2021
- Full Text
- View/download PDF
16. CDK2 phosphorylation of Werner protein (WRN) contributes to WRN's DNA double-strand break repair pathway choice.
- Author
-
Lee JH, Shamanna RA, Kulikowicz T, Borhan Fakouri N, Kim EW, Christiansen LS, Croteau DL, and Bohr VA
- Subjects
- Cell Cycle genetics, Cell Line, Tumor, Cyclin-Dependent Kinase 2 genetics, DNA metabolism, HEK293 Cells, Humans, Phosphorylation genetics, Replication Protein A metabolism, Serine metabolism, Transfection, Werner Syndrome genetics, Werner Syndrome metabolism, Werner Syndrome Helicase genetics, Cyclin-Dependent Kinase 2 metabolism, DNA Breaks, Double-Stranded radiation effects, DNA End-Joining Repair genetics, Homologous Recombination, Signal Transduction genetics, Werner Syndrome Helicase metabolism
- Abstract
Werner syndrome (WS) is an accelerated aging disorder characterized by genomic instability, which is caused by WRN protein deficiency. WRN participates in DNA metabolism including DNA repair. In a previous report, we showed that WRN protein is recruited to laser-induced DNA double-strand break (DSB) sites during various stages of the cell cycle with similar intensities, supporting that WRN participates in both non-homologous end joining (NHEJ) and homologous recombination (HR). Here, we demonstrate that the phosphorylation of WRN by CDK2 on serine residue 426 is critical for WRN to make its DSB repair pathway choice between NHEJ and HR. Cells expressing WRN engineered to mimic the unphosphorylated or phosphorylation state at serine 426 showed abnormal DSB recruitment, altered RPA interaction, strand annealing, and DSB repair activities. The CDK2 phosphorylation on serine 426 stabilizes WRN's affinity for RPA, likely increasing its long-range resection at the end of DNA strands, which is a crucial step for HR. Collectively, the data shown here demonstrate that a CDK2-dependent phosphorylation of WRN regulates DSB repair pathway choice and cell cycle participation., (Published 2021. This article is a U.S. Government work and is in the public domain in the USA.)
- Published
- 2021
- Full Text
- View/download PDF
17. NAD + augmentation with nicotinamide riboside improves lymphoid potential of Atm -/- and old mice HSCs.
- Author
-
Zong L, Tanaka-Yano M, Park B, Yanai H, Turhan FT, Croteau DL, Tian J, Fang EF, Bohr VA, and Beerman I
- Abstract
NAD
+ supplementation has significant benefits in compromised settings, acting largely through improved mitochondrial function and DNA repair. Elevating NAD+ to physiological levels has been shown to improve the function of some adult stem cells, with implications that these changes will lead to sustained improvement of the tissue or system. Here, we examined the effect of elevating NAD+ levels in models with reduced hematopoietic stem cell (HSC) potential, ATM-deficient and aged WT mice, and showed that supplementation of nicotinamide riboside (NR), a NAD+ precursor, improved lymphoid lineage potential during supplementation. In aged mice, this improved lymphoid potential was maintained in competitive transplants and was associated with transcriptional repression of myeloid gene signatures in stem and lineage-committed progenitor cells after NR treatment. However, the altered transcriptional priming of the stem cells toward lymphoid lineages was not sustained in the aged mice after NR removal. These data characterize significant alterations to the lineage potential of functionally compromised HSCs after short-term exposure to NR treatment., (© 2021. This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply.)- Published
- 2021
- Full Text
- View/download PDF
18. NAD + supplementation reduces neuroinflammation and cell senescence in a transgenic mouse model of Alzheimer's disease via cGAS-STING.
- Author
-
Hou Y, Wei Y, Lautrup S, Yang B, Wang Y, Cordonnier S, Mattson MP, Croteau DL, and Bohr VA
- Subjects
- Animals, Humans, Membrane Proteins genetics, Mice, Mice, Transgenic, Neuroinflammatory Diseases etiology, Neuroinflammatory Diseases pathology, Niacinamide administration & dosage, Niacinamide analogs & derivatives, Nucleotidyltransferases genetics, Pyridinium Compounds administration & dosage, Alzheimer Disease complications, Cellular Senescence, Dietary Supplements, Membrane Proteins metabolism, NAD administration & dosage, Neuroinflammatory Diseases drug therapy, Nucleotidyltransferases metabolism
- Abstract
Alzheimer's disease (AD) is a progressive and fatal neurodegenerative disorder. Impaired neuronal bioenergetics and neuroinflammation are thought to play key roles in the progression of AD, but their interplay is not clear. Nicotinamide adenine dinucleotide (NAD
+ ) is an important metabolite in all human cells in which it is pivotal for multiple processes including DNA repair and mitophagy, both of which are impaired in AD neurons. Here, we report that levels of NAD+ are reduced and markers of inflammation increased in the brains of APP/PS1 mutant transgenic mice with beta-amyloid pathology. Treatment of APP/PS1 mutant mice with the NAD+ precursor nicotinamide riboside (NR) for 5 mo increased brain NAD+ levels, reduced expression of proinflammatory cytokines, and decreased activation of microglia and astrocytes. NR treatment also reduced NLRP3 inflammasome expression, DNA damage, apoptosis, and cellular senescence in the AD mouse brains. Activation of cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) are associated with DNA damage and senescence. cGAS-STING elevation was observed in the AD mice and normalized by NR treatment. Cell culture experiments using microglia suggested that the beneficial effects of NR are, in part, through a cGAS-STING-dependent pathway. Levels of ectopic (cytoplasmic) DNA were increased in APP/PS1 mutant mice and human AD fibroblasts and down-regulated by NR. NR treatment induced mitophagy and improved cognitive and synaptic functions in APP/PS1 mutant mice. Our findings suggest a role for NAD+ depletion-mediated activation of cGAS-STING in neuroinflammation and cellular senescence in AD., Competing Interests: Competing interest statement: V.A.B. has a Cooperative Research and Development Agreement (CRADA) with Chromadex Corporation but receives no personal benefits.- Published
- 2021
- Full Text
- View/download PDF
19. Olfactory dysfunction in aging and neurodegenerative diseases.
- Author
-
Dan X, Wechter N, Gray S, Mohanty JG, Croteau DL, and Bohr VA
- Subjects
- Aging, Animals, Humans, Smell, Alzheimer Disease diagnosis, Neurodegenerative Diseases, Olfaction Disorders diagnosis, Parkinson Disease
- Abstract
Alterations in olfactory functions are proposed to be early biomarkers for neurodegeneration. Many neurodegenerative diseases are age-related, including two of the most common, Parkinson's disease (PD) and Alzheimer's disease (AD). The establishment of biomarkers that promote early risk identification is critical for the implementation of early treatment to postpone or avert pathological development. Olfactory dysfunction (OD) is seen in 90% of early-stage PD patients and 85% of patients with early-stage AD, which makes it an attractive biomarker for early diagnosis of these diseases. Here, we systematically review widely applied smelling tests available for humans as well as olfaction assessments performed in some animal models and the relationships between OD and normal aging, PD, AD, and other conditions. The utility of OD as a biomarker for neurodegenerative disease diagnosis and future research directions are also discussed., (Published by Elsevier B.V.)
- Published
- 2021
- Full Text
- View/download PDF
20. Skin Abnormalities in Disorders with DNA Repair Defects, Premature Aging, and Mitochondrial Dysfunction.
- Author
-
Hussain M, Krishnamurthy S, Patel J, Kim E, Baptiste BA, Croteau DL, and Bohr VA
- Subjects
- Aging, Premature genetics, Aging, Premature pathology, Animals, Apoptosis genetics, Cockayne Syndrome complications, Cockayne Syndrome genetics, Cockayne Syndrome pathology, Disease Models, Animal, Energy Metabolism genetics, Humans, Multiple Endocrine Neoplasia Type 1 complications, Multiple Endocrine Neoplasia Type 1 genetics, Multiple Endocrine Neoplasia Type 1 pathology, Rothmund-Thomson Syndrome complications, Rothmund-Thomson Syndrome genetics, Rothmund-Thomson Syndrome pathology, Skin cytology, Skin Diseases pathology, Aging, Premature complications, DNA Repair, Mitochondria pathology, Skin pathology, Skin Diseases genetics
- Abstract
Defects in DNA repair pathways and alterations of mitochondrial energy metabolism have been reported in multiple skin disorders. More than 10% of patients with primary mitochondrial dysfunction exhibit dermatological features including rashes and hair and pigmentation abnormalities. Accumulation of oxidative DNA damage and dysfunctional mitochondria affect cellular homeostasis leading to increased apoptosis. Emerging evidence demonstrates that genetic disorders of premature aging that alter DNA repair pathways and cause mitochondrial dysfunction, such as Rothmund-Thomson syndrome, Werner syndrome, and Cockayne syndrome, also exhibit skin disease. This article summarizes recent advances in the research pertaining to these syndromes and molecular mechanisms underlying their skin pathologies., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
21. NAD + supplementation prevents STING-induced senescence in ataxia telangiectasia by improving mitophagy.
- Author
-
Yang B, Dan X, Hou Y, Lee JH, Wechter N, Krishnamurthy S, Kimura R, Babbar M, Demarest T, McDevitt R, Zhang S, Zhang Y, Mattson MP, Croteau DL, and Bohr VA
- Subjects
- Animals, Ataxia Telangiectasia genetics, Ataxia Telangiectasia Mutated Proteins genetics, Ataxia Telangiectasia Mutated Proteins metabolism, Case-Control Studies, Cell Line, Tumor, Disease Models, Animal, Female, Fibroblasts drug effects, Fibroblasts metabolism, Humans, Male, Membrane Proteins genetics, Mice, Mice, Knockout, Mitochondria metabolism, Mitophagy genetics, Neurons drug effects, Neurons metabolism, Niacinamide administration & dosage, Rats, Rats, Sprague-Dawley, Signal Transduction genetics, Transfection, Treatment Outcome, Ataxia Telangiectasia diet therapy, Ataxia Telangiectasia metabolism, Dietary Supplements, Membrane Proteins metabolism, Mitophagy drug effects, NAD metabolism, Niacinamide analogs & derivatives, Pyridinium Compounds administration & dosage, Senescence-Associated Secretory Phenotype genetics, Signal Transduction drug effects
- Abstract
Senescence phenotypes and mitochondrial dysfunction are implicated in aging and in premature aging diseases, including ataxia telangiectasia (A-T). Loss of mitochondrial function can drive age-related decline in the brain, but little is known about whether improving mitochondrial homeostasis alleviates senescence phenotypes. We demonstrate here that mitochondrial dysfunction and cellular senescence with a senescence-associated secretory phenotype (SASP) occur in A-T patient fibroblasts, and in ATM-deficient cells and mice. Senescence is mediated by stimulator of interferon genes (STING) and involves ectopic cytoplasmic DNA. We further show that boosting intracellular NAD
+ levels with nicotinamide riboside (NR) prevents senescence and SASP by promoting mitophagy in a PINK1-dependent manner. NR treatment also prevents neurodegeneration, suppresses senescence and neuroinflammation, and improves motor function in Atm-/- mice. Our findings suggest a central role for mitochondrial dysfunction-induced senescence in A-T pathogenesis, and that enhancing mitophagy as a potential therapeutic intervention., (Published 2021. This article is a U.S. Government work and is in the public domain in the USA. Aging Cell published by Anatomical Society and John Wiley & Sons Ltd.)- Published
- 2021
- Full Text
- View/download PDF
22. Self-assembly of multi-component mitochondrial nucleoids via phase separation.
- Author
-
Feric M, Demarest TG, Tian J, Croteau DL, Bohr VA, and Misteli T
- Subjects
- Cell Line, Child, Child, Preschool, DNA-Binding Proteins metabolism, HEK293 Cells, Humans, Mitochondrial Proteins metabolism, Progeria genetics, Transcription Factors metabolism, DNA, Mitochondrial genetics, Genome, Mitochondrial genetics, Mitochondria genetics, Progeria pathology
- Abstract
Mitochondria contain an autonomous and spatially segregated genome. The organizational unit of their genome is the nucleoid, which consists of mitochondrial DNA (mtDNA) and associated architectural proteins. Here, we show that phase separation is the primary physical mechanism for assembly and size control of the mitochondrial nucleoid (mt-nucleoid). The major mtDNA-binding protein TFAM spontaneously phase separates in vitro via weak, multivalent interactions into droplets with slow internal dynamics. TFAM and mtDNA form heterogenous, viscoelastic structures in vitro, which recapitulate the dynamics and behavior of mt-nucleoids in vivo. Mt-nucleoids coalesce into larger droplets in response to various forms of cellular stress, as evidenced by the enlarged and transcriptionally active nucleoids in mitochondria from patients with the premature aging disorder Hutchinson-Gilford Progeria Syndrome (HGPS). Our results point to phase separation as an evolutionarily conserved mechanism of genome organization., (© 2021 This article is a U.S. Government work and is in the public domain in the USA.)
- Published
- 2021
- Full Text
- View/download PDF
23. DNA polymerase β outperforms DNA polymerase γ in key mitochondrial base excision repair activities.
- Author
-
Baptiste BA, Baringer SL, Kulikowicz T, Sommers JA, Croteau DL, Brosh RM Jr, and Bohr VA
- Subjects
- Animals, DNA Damage, Mice, Mitochondria genetics, DNA Polymerase beta metabolism, DNA Polymerase gamma metabolism, DNA Repair, DNA, Mitochondrial metabolism, Mitochondria metabolism
- Abstract
DNA polymerase beta (POLβ), well known for its role in nuclear DNA base excision repair (BER), has been shown to be present in the mitochondria of several different cell types. Here we present a side-by-side comparison of BER activities of POLβ and POLγ, the mitochondrial replicative polymerase, previously thought to be the only mitochondrial polymerase. We find that POLβ is significantly more proficient at single-nucleotide gap filling, both in substrates with ends that require polymerase processing, and those that do not. We also show that POLβ has a helicase-independent functional interaction with the mitochondrial helicase, TWINKLE. This interaction stimulates strand-displacement synthesis, but not single-nucleotide gap filling. Importantly, we find that purified mitochondrial extracts from cells lacking POLβ are severely deficient in processing BER intermediates, suggesting that mitochondrially localized DNA POLβ may be critical for cells with high energetic demands that produce greater levels of oxidative stress and therefore depend upon efficient BER for mitochondrial health., (Published by Elsevier B.V.)
- Published
- 2021
- Full Text
- View/download PDF
24. DNA damage and mitochondria in cancer and aging.
- Author
-
Patel J, Baptiste BA, Kim E, Hussain M, Croteau DL, and Bohr VA
- Subjects
- Aging genetics, Animals, Humans, Neoplasms genetics, Signal Transduction, Telomere, Aging pathology, DNA Damage, DNA Repair, Mitochondria genetics, Neoplasms pathology
- Abstract
Age and DNA repair deficiencies are strong risk factors for developing cancer. This is reflected in the comorbidity of cancer with premature aging diseases associated with DNA damage repair deficiencies. Recent research has suggested that DNA damage accumulation, telomere dysfunction and the accompanying mitochondrial dysfunction exacerbate the aging process and may increase the risk of cancer development. Thus, an area of interest in both cancer and aging research is the elucidation of the dynamic crosstalk between the nucleus and the mitochondria. In this review, we discuss current research on aging and cancer with specific focus on the role of mitochondrial dysfunction in cancer and aging as well as how nuclear to mitochondrial DNA damage signaling may be a driving factor in the increased cancer incidence with aging. We suggest that therapeutic interventions aimed at the induction of autophagy and mediation of nuclear to mitochondrial signaling may provide a mechanism for healthier aging and reduced tumorigenesis., (Published by Oxford University Press 2020.)
- Published
- 2020
- Full Text
- View/download PDF
25. Cockayne syndrome proteins CSA and CSB maintain mitochondrial homeostasis through NAD + signaling.
- Author
-
Okur MN, Fang EF, Fivenson EM, Tiwari V, Croteau DL, and Bohr VA
- Subjects
- 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, Cockayne Syndrome metabolism, DNA Helicases metabolism, DNA Repair Enzymes metabolism, Mitochondria metabolism, NAD metabolism, Poly-ADP-Ribose Binding Proteins metabolism, Transcription Factors metabolism
- 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., (Published 2020. This article is a U.S. Government work and is in the public domain in the USA.)- Published
- 2020
- Full Text
- View/download PDF
26. Spatial Transcriptomics Reveals Genes Associated with Dysregulated Mitochondrial Functions and Stress Signaling in Alzheimer Disease.
- Author
-
Navarro JF, Croteau DL, Jurek A, Andrusivova Z, Yang B, Wang Y, Ogedegbe B, Riaz T, Støen M, Desler C, Rasmussen LJ, Tønjum T, Galas MC, Lundeberg J, and Bohr VA
- Abstract
Alzheimer disease (AD) is a devastating neurological disease associated with progressive loss of mental skills and cognitive and physical functions whose etiology is not completely understood. Here, our goal was to simultaneously uncover novel and known molecular targets in the structured layers of the hippocampus and olfactory bulbs that may contribute to early hippocampal synaptic deficits and olfactory dysfunction in AD mice. Spatially resolved transcriptomics was used to identify high-confidence genes that were differentially regulated in AD mice relative to controls. A diverse set of genes that modulate stress responses and transcription were predominant in both hippocampi and olfactory bulbs. Notably, we identify Bok, implicated in mitochondrial physiology and cell death, as a spatially downregulated gene in the hippocampus of mouse and human AD brains. In summary, we provide a rich resource of spatially differentially expressed genes, which may contribute to understanding AD pathology., Competing Interests: J.L. and J.F.N are scientific advisors at 10x Genomics Inc, which provides commercial barcoded arrays. All other authors declare no competing interests.
- Published
- 2020
- Full Text
- View/download PDF
27. Heterochromatin: an epigenetic point of view in aging.
- Author
-
Lee JH, Kim EW, Croteau DL, and Bohr VA
- Subjects
- Age Factors, Animals, Chromatin genetics, Chromatin metabolism, Gene Expression Regulation, Heterochromatin metabolism, Histones metabolism, Humans, Longevity, Aging genetics, Epigenesis, Genetic, Epigenomics methods, Heterochromatin genetics
- Abstract
Aging is an inevitable process of life. Defined by progressive physiological and functional loss of tissues and organs, aging increases the risk of mortality for the organism. The aging process is affected by various factors, including genetic and epigenetic ones. Here, we review the chromatin-specific epigenetic changes that occur during normal (chronological) aging and in premature aging diseases. Taking advantage of the reversible nature of epigenetic modifications, we will also discuss possible lifespan expansion strategies through epigenetic modulation, which was considered irreversible until recently.
- Published
- 2020
- Full Text
- View/download PDF
28. DNA damage invokes mitophagy through a pathway involving Spata18.
- Author
-
Dan X, Babbar M, Moore A, Wechter N, Tian J, Mohanty JG, Croteau DL, and Bohr VA
- Subjects
- Animals, Caenorhabditis elegans genetics, Cell Line, Cell Proliferation genetics, DNA Damage genetics, DNA Repair genetics, Fibroblasts metabolism, Humans, Mice, Mitochondria genetics, Calcium metabolism, Mitochondrial Proteins genetics, Mitophagy genetics, Neurons metabolism
- Abstract
Mitochondria are vital for cellular energy supply and intracellular signaling after stress. Here, we aimed to investigate how mitochondria respond to acute DNA damage with respect to mitophagy, which is an important mitochondrial quality control process. Our results show that mitophagy increases after DNA damage in primary fibroblasts, murine neurons and Caenorhabditis elegans neurons. Our results indicate that modulation of mitophagy after DNA damage is independent of the type of DNA damage stimuli used and that the protein Spata18 is an important player in this process. Knockdown of Spata18 suppresses mitophagy, disturbs mitochondrial Ca2+ homeostasis, affects ATP production, and attenuates DNA repair. Importantly, mitophagy after DNA damage is a vital cellular response to maintain mitochondrial functions and DNA repair., (Published by Oxford University Press on behalf of Nucleic Acids Research 2020.)
- Published
- 2020
- Full Text
- View/download PDF
29. Interaction between RECQL4 and OGG1 promotes repair of oxidative base lesion 8-oxoG and is regulated by SIRT1 deacetylase.
- Author
-
Duan S, Han X, Akbari M, Croteau DL, Rasmussen LJ, and Bohr VA
- Subjects
- Acetylation, Cell Line, Tumor, Guanosine analogs & derivatives, Guanosine genetics, HEK293 Cells, Humans, Oxidative Stress, Protein Binding, DNA Glycosylases metabolism, DNA Repair, RecQ Helicases metabolism, Sirtuin 1 metabolism
- Abstract
OGG1 initiated base excision repair (BER) is the major pathway for repair of oxidative DNA base damage 8-oxoguanine (8-oxoG). Here, we report that RECQL4 DNA helicase, deficient in the cancer-prone and premature aging Rothmund-Thomson syndrome, physically and functionally interacts with OGG1. RECQL4 promotes catalytic activity of OGG1 and RECQL4 deficiency results in defective 8-oxoG repair and increased genomic 8-oxoG. Furthermore, we show that acute oxidative stress leads to increased RECQL4 acetylation and its interaction with OGG1. The NAD+-dependent protein SIRT1 deacetylates RECQL4 in vitro and in cells thereby controlling the interaction between OGG1 and RECQL4 after DNA repair and maintaining RECQL4 in a low acetylated state. Collectively, we find that RECQL4 is involved in 8-oxoG repair through interaction with OGG1, and that SIRT1 indirectly modulates BER of 8-oxoG by controlling RECQL4-OGG1 interaction., (© The Author(s) 2020. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2020
- Full Text
- View/download PDF
30. Biological sex and DNA repair deficiency drive Alzheimer's disease via systemic metabolic remodeling and brain mitochondrial dysfunction.
- Author
-
Demarest TG, Varma VR, Estrada D, Babbar M, Basu S, Mahajan UV, Moaddel R, Croteau DL, Thambisetty M, Mattson MP, and Bohr VA
- Subjects
- Alzheimer Disease pathology, Animals, Brain pathology, Caenorhabditis elegans, DNA Repair-Deficiency Disorders pathology, Energy Metabolism physiology, Female, Glucose metabolism, Humans, Lipid Metabolism physiology, Male, Mice, Mitochondria pathology, Alzheimer Disease metabolism, Brain metabolism, DNA Repair-Deficiency Disorders metabolism, Mitochondria metabolism, Sex Characteristics
- Abstract
Alzheimer's disease (AD) is an incurable neurodegenerative disease that is more prevalent in women. The increased risk of AD in women is not well understood. It is well established that there are sex differences in metabolism and that metabolic alterations are an early component of AD. We utilized a cross-species approach to evaluate conserved metabolic alterations in the serum and brain of human AD subjects, two AD mouse models, a human cell line, and two Caenorhabditis elegans AD strains. We found a mitochondrial complex I-specific impairment in cortical synaptic brain mitochondria in female, but not male, AD mice. In the hippocampus, Polβ haploinsufficiency caused synaptic complex I impairment in male and female mice, demonstrating the critical role of DNA repair in mitochondrial function. In non-synaptic, glial-enriched, mitochondria from the cortex and hippocampus, complex II-dependent respiration increased in female, but not male, AD mice. These results suggested a glial upregulation of fatty acid metabolism to compensate for neuronal glucose hypometabolism in AD. Using an unbiased metabolomics approach, we consistently observed evidence of systemic and brain metabolic remodeling with a shift from glucose to lipid metabolism in humans with AD, and in AD mice. We determined that this metabolic shift is necessary for cellular and organismal survival in C. elegans, and human cell culture AD models. We observed sex-specific, systemic, and brain metabolic alterations in humans with AD, and that these metabolite changes significantly correlate with amyloid and tau pathology. Among the most significant metabolite changes was the accumulation of glucose-6-phosphate in AD, an inhibitor of hexokinase and rate-limiting metabolite for the pentose phosphate pathway (PPP). Overall, we identified novel mechanisms of glycolysis inhibition, PPP, and tricarboxylic acid cycle impairment, and a neuroprotective augmentation of lipid metabolism in AD. These findings support a sex-targeted metabolism-modifying strategy to prevent and treat AD.
- Published
- 2020
- Full Text
- View/download PDF
31. Cockayne syndrome group A and B proteins function in rRNA transcription through nucleolin regulation.
- Author
-
Okur MN, Lee JH, Osmani W, Kimura R, Demarest TG, Croteau DL, and Bohr VA
- Subjects
- Cell Line, DNA, Ribosomal genetics, Humans, Models, Biological, Phosphoproteins metabolism, Protein Binding, RNA-Binding Proteins metabolism, Nucleolin, 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
- 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., (Published by Oxford University Press on behalf of Nucleic Acids Research 2020.)
- Published
- 2020
- Full Text
- View/download PDF
32. Hippocampal tau oligomerization early in tau pathology coincides with a transient alteration of mitochondrial homeostasis and DNA repair in a mouse model of tauopathy.
- Author
-
Zheng J, Akbari M, Schirmer C, Reynaert ML, Loyens A, Lefebvre B, Buée L, Croteau DL, Galas MC, and Bohr VA
- Subjects
- Adenine Nucleotide Translocator 1 metabolism, Aged, Animals, DNA Damage, DNA Polymerase beta metabolism, DNA Repair, Disease Models, Animal, Dynamins metabolism, Frontal Lobe cytology, Frontal Lobe metabolism, Hippocampus cytology, Homeostasis, Humans, Male, Mice, Transgenic, Microscopy, Electron, Transmission, Microscopy, Immunoelectron, Middle Aged, Mitochondria ultrastructure, Neurofibrillary Tangles, Neurons ultrastructure, Protein Kinases metabolism, Sirtuin 3 metabolism, tau Proteins genetics, Alzheimer Disease metabolism, Hippocampus metabolism, Mitochondria metabolism, Neurons metabolism, Oxidative Stress, tau Proteins metabolism
- Abstract
Insoluble intracellular aggregation of tau proteins into filaments and neurodegeneration are histopathological hallmarks of Alzheimer disease (AD) and other tauopathies. Recently, prefibrillar, soluble, oligomeric tau intermediates have emerged as relevant pathological tau species; however, the molecular mechanisms of neuronal responses to tau oligomers are not fully understood. Here, we show that hippocampal neurons in six-month-old transgenic mouse model of tauopathy, THY-Tau22, are enriched with oligomeric tau, contain elongated mitochondria, and display cellular stress, but no overt cytotoxicity compared to the control mice. The levels of several key mitochondrial proteins were markedly different between the THY-Tau22 and control mice hippocampi including the mitochondrial SIRT3, PINK1, ANT1 and the fission protein DRP1. DNA base excision repair (BER) is the primary defense system against oxidative DNA damage and it was elevated in six-month-old transgenic mice. DNA polymerase β, the key BER DNA polymerase, was enriched in the cytoplasm of hippocampal neurons in six-month-old transgenic mice and localized with and within mitochondria. Polβ also co-localized with mitochondria in human AD brains in neurons containing oligomeric tau. Most of these altered mitochondrial and DNA repair events were specific to the transgenic mice at 6 months of age and were not different from control mice at 12 months of age when tau pathology reaches its maximum and oligomeric forms of tau are no longer detectable. In summary, our data suggests that we have identified key cellular stress responses at early stages of tau pathology to preserve neuronal integrity and to promote survival. To our knowledge, this work provides the first description of multiple stress responses involving mitochondrial homeostasis and BER early during the progression of tau pathology, and represents an important advance in the etiopathogenesis of tauopathies.
- Published
- 2020
- Full Text
- View/download PDF
33. Mitophagy and DNA damage signaling in human aging.
- Author
-
Babbar M, Basu S, Yang B, Croteau DL, and Bohr VA
- Subjects
- Aging genetics, DNA Repair, Humans, Signal Transduction, Aging physiology, DNA Damage, Mitophagy physiology
- Abstract
Aging is associated with multiple human pathologies. In the past few years mitochondrial homeostasis has been well correlated with age-related disorders and longevity. Mitochondrial homeostasis involves generation, biogenesis and removal of dysfunctional mitochondria via mitophagy. Mitophagy is regulated by various mitochondrial and extra-mitochondrial factors including morphology, oxidative stress and DNA damage. For decades, DNA damage and inefficient DNA repair have been considered as major determinants for age-related disorders. Although defects in DNA damage recognition and repair and mitophagy are well documented to be major factors in age-associated diseases, interactivity between these is poorly understood. Mitophagy efficiency decreases with age leading to accumulation of dysfunctional mitochondria enhancing the severity of age-related disorders including neurodegenerative diseases, inflammatory diseases, cancer, diabetes and many more. Therefore, mitophagy is being targeted for intervention in age-associated disorders. NAD
+ supplementation has emerged as one intervention to target both defective DNA repair and mitophagy. In this review, we discuss the molecular signaling pathways involved in regulation of DNA damage and repair and of mitophagy, and we highlight the opportunities for clinical interventions targeting these processes to improve the quality of life during aging., (Published by Elsevier B.V.)- Published
- 2020
- Full Text
- View/download PDF
34. Short-term NAD + supplementation prevents hearing loss in mouse models of Cockayne syndrome.
- Author
-
Okur MN, Mao B, Kimura R, Haraczy S, Fitzgerald T, Edwards-Hollingsworth K, Tian J, Osmani W, Croteau DL, Kelley MW, and Bohr VA
- 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 CSBm/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 CSBm/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 CSBm/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 CSBm/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 CSBm/m mutant cochleae. These findings provide valuable insight into the mechanism of CS- and ARHL-associated hearing loss, and suggest a possible intervention., Competing Interests: Competing interestsV.A.B. receives nicotinamide riboside from Chromadex Corp., (© This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply 2020.)- Published
- 2020
- Full Text
- View/download PDF
35. NAD + augmentation restores mitophagy and limits accelerated aging in Werner syndrome.
- Author
-
Fang EF, Hou Y, Lautrup S, Jensen MB, Yang B, SenGupta T, Caponio D, Khezri R, Demarest TG, Aman Y, Figueroa D, Morevati M, Lee HJ, Kato H, Kassahun H, Lee JH, Filippelli D, Okur MN, Mangerich A, Croteau DL, Maezawa Y, Lyssiotis CA, Tao J, Yokote K, Rusten TE, Mattson MP, Jasper H, Nilsen H, and Bohr VA
- Subjects
- 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, Aging, Premature metabolism, Mitophagy, NAD metabolism, Werner Syndrome metabolism, Werner Syndrome Helicase metabolism
- 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.- Published
- 2019
- Full Text
- View/download PDF
36. Ageing as a risk factor for neurodegenerative disease.
- Author
-
Hou Y, Dan X, Babbar M, Wei Y, Hasselbalch SG, Croteau DL, and Bohr VA
- Subjects
- Aging pathology, Brain pathology, DNA Damage physiology, Epigenesis, Genetic physiology, Humans, Mitophagy physiology, Neurodegenerative Diseases pathology, Risk Factors, Aging genetics, Aging metabolism, Brain metabolism, Neurodegenerative Diseases genetics, Neurodegenerative Diseases metabolism
- Abstract
Ageing is the primary risk factor for most neurodegenerative diseases, including Alzheimer disease (AD) and Parkinson disease (PD). One in ten individuals aged ≥65 years has AD and its prevalence continues to increase with increasing age. Few or no effective treatments are available for ageing-related neurodegenerative diseases, which tend to progress in an irreversible manner and are associated with large socioeconomic and personal costs. This Review discusses the pathogenesis of AD, PD and other neurodegenerative diseases, and describes their associations with the nine biological hallmarks of ageing: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, mitochondrial dysfunction, cellular senescence, deregulated nutrient sensing, stem cell exhaustion and altered intercellular communication. The central biological mechanisms of ageing and their potential as targets of novel therapies for neurodegenerative diseases are also discussed, with potential therapies including NAD
+ precursors, mitophagy inducers and inhibitors of cellular senescence.- Published
- 2019
- Full Text
- View/download PDF
37. Cockayne syndrome group B deficiency reduces H3K9me3 chromatin remodeler SETDB1 and exacerbates cellular aging.
- Author
-
Lee JH, Demarest TG, Babbar M, Kim EW, Okur MN, De S, Croteau DL, and Bohr VA
- Subjects
- 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, 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
- 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., (Published by Oxford University Press on behalf of Nucleic Acids Research 2019.)
- Published
- 2019
- Full Text
- View/download PDF
38. NEIL1 stimulates neurogenesis and suppresses neuroinflammation after stress.
- Author
-
Yang B, Figueroa DM, Hou Y, Babbar M, Baringer SL, Croteau DL, and Bohr VA
- Subjects
- Aging, Animals, Apoptosis, Cell Proliferation, Central Nervous System metabolism, DNA Damage, DNA Glycosylases genetics, DNA Repair, Fear, Gamma Rays, Gene Expression Profiling, Hippocampus metabolism, Male, Maze Learning, Mice, Mice, Inbred C57BL, Mice, Knockout, Neural Stem Cells cytology, Oligonucleotide Array Sequence Analysis, RNA analysis, Behavior, Animal, DNA Glycosylases metabolism, Inflammation, Neurodegenerative Diseases metabolism, Stress, Psychological metabolism
- Abstract
Cellular exposure to ionizing radiation leads to oxidatively generated DNA damage, which has been implicated in neurodegenerative diseases. DNA damage is repaired by the evolutionarily conserved base excision repair (BER) system. Exposure of mice to ionizing radiation affects neurogenesis and neuroinflammation. However, the consequences of deficient DNA repair on adult neurogenesis and neuroinflammation are poorly understood despite their potential relevance for homeostasis. We previously reported that loss of NEIL1, an important DNA glycosylase involved in BER, is associated with deficiencies in spatial memory, olfaction, and protection against ischemic stroke in mice. Here, we show that Neil1
-/- mice display an anxiety-mediated behavior in the open field test, a deficient recognitive memory in novel object recognition and increased neuroinflammatory response under basal conditions. Further, mice lacking NEIL1 have decreased neurogenesis and deficient resolution of neuroinflammation following gamma irradiation (IR)-induced stress compared to WT mice. Neil1-/- IR-exposed mice also exhibit increased DNA damage and apoptosis in the hippocampus. Interestingly, behavioral tests two weeks after IR showed impaired stress response in the Neil1-/- mice. Our data indicate that NEIL1 plays an important role in adult neurogenesis and in the resolution of neuroinflammation., (Copyright © 2019. Published by Elsevier Inc.)- Published
- 2019
- Full Text
- View/download PDF
39. Diminished OPA1 expression and impaired mitochondrial morphology and homeostasis in Aprataxin-deficient cells.
- Author
-
Zheng J, Croteau DL, Bohr VA, and Akbari M
- Subjects
- Cell Line, Transformed, Cell Line, Tumor, DNA-Binding Proteins deficiency, Electron Transport Chain Complex Proteins genetics, Electron Transport Chain Complex Proteins metabolism, GTP Phosphohydrolases deficiency, Gene Expression Profiling, Gene Expression Regulation, Homeostasis genetics, Humans, Lymphocytes metabolism, Lymphocytes pathology, Mitochondria metabolism, Mitochondria ultrastructure, Nuclear Proteins deficiency, Oligonucleotide Array Sequence Analysis, Osteoblasts metabolism, Osteoblasts pathology, Oxidative Phosphorylation, Signal Transduction, Spinocerebellar Ataxias genetics, Spinocerebellar Ataxias metabolism, Spinocerebellar Ataxias pathology, DNA-Binding Proteins genetics, GTP Phosphohydrolases genetics, Mitochondria genetics, Mitophagy genetics, Nuclear Proteins genetics, Spinocerebellar Ataxias congenital
- Abstract
Ataxia with oculomotor apraxia type 1 (AOA1) is an early onset progressive spinocerebellar ataxia caused by mutation in aprataxin (APTX). APTX removes 5'-AMP groups from DNA, a product of abortive ligation during DNA repair and replication. APTX deficiency has been suggested to compromise mitochondrial function; however, a detailed characterization of mitochondrial homeostasis in APTX-deficient cells is not available. Here, we show that cells lacking APTX undergo mitochondrial stress and display significant changes in the expression of the mitochondrial inner membrane fusion protein optic atrophy type 1, and components of the oxidative phosphorylation complexes. At the cellular level, APTX deficiency impairs mitochondrial morphology and network formation, and autophagic removal of damaged mitochondria by mitophagy. Thus, our results show that aberrant mitochondrial function is a key component of AOA1 pathology. This work corroborates the emerging evidence that impaired mitochondrial function is a characteristic of an increasing number of genetically diverse neurodegenerative disorders., (Published by Oxford University Press on behalf of Nucleic Acids Research 2019.)
- Published
- 2019
- Full Text
- View/download PDF
40. Mitophagy inhibits amyloid-β and tau pathology and reverses cognitive deficits in models of Alzheimer's disease.
- Author
-
Fang EF, Hou Y, Palikaras K, Adriaanse BA, Kerr JS, Yang B, Lautrup S, Hasan-Olive MM, Caponio D, Dan X, Rocktäschel P, Croteau DL, Akbari M, Greig NH, Fladby T, Nilsen H, Cader MZ, Mattson MP, Tavernarakis N, and Bohr VA
- Subjects
- Alzheimer Disease pathology, Alzheimer Disease psychology, Animals, Animals, Genetically Modified, Caenorhabditis elegans, Disease Models, Animal, Female, Induced Pluripotent Stem Cells, Male, Memory, Mice, Neural Stem Cells, Alzheimer Disease metabolism, Amyloid beta-Peptides metabolism, Hippocampus metabolism, Hippocampus pathology, Mitophagy, Neurons metabolism, Neurons pathology
- Abstract
Accumulation of damaged mitochondria is a hallmark of aging and age-related neurodegeneration, including Alzheimer's disease (AD). The molecular mechanisms of impaired mitochondrial homeostasis in AD are being investigated. Here we provide evidence that mitophagy is impaired in the hippocampus of AD patients, in induced pluripotent stem cell-derived human AD neurons, and in animal AD models. In both amyloid-β (Aβ) and tau Caenorhabditis elegans models of AD, mitophagy stimulation (through NAD
+ supplementation, urolithin A, and actinonin) reverses memory impairment through PINK-1 (PTEN-induced kinase-1)-, PDR-1 (Parkinson's disease-related-1; parkin)-, or DCT-1 (DAF-16/FOXO-controlled germline-tumor affecting-1)-dependent pathways. Mitophagy diminishes insoluble Aβ1-42 and Aβ1-40 and prevents cognitive impairment in an APP/PS1 mouse model through microglial phagocytosis of extracellular Aβ plaques and suppression of neuroinflammation. Mitophagy enhancement abolishes AD-related tau hyperphosphorylation in human neuronal cells and reverses memory impairment in transgenic tau nematodes and mice. Our findings suggest that impaired removal of defective mitochondria is a pivotal event in AD pathogenesis and that mitophagy represents a potential therapeutic intervention.- Published
- 2019
- Full Text
- View/download PDF
41. Toward understanding genomic instability, mitochondrial dysfunction and aging.
- Author
-
Fakouri NB, Hou Y, Demarest TG, Christiansen LS, Okur MN, Mohanty JG, Croteau DL, and Bohr VA
- Subjects
- Aging metabolism, Animals, Energy Metabolism, Humans, Mitochondria metabolism, Mitophagy, Poly(ADP-ribose) Polymerases metabolism, Aging pathology, Genomic Instability, Homeostasis, Mitochondria pathology, Oxidative Stress
- 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., (Published 2018. This article is a U.S. Government work and is in the public domain in the USA.)
- Published
- 2019
- Full Text
- View/download PDF
42. A high-throughput screen to identify novel small molecule inhibitors of the Werner Syndrome Helicase-Nuclease (WRN).
- Author
-
Sommers JA, Kulikowicz T, Croteau DL, Dexheimer T, Dorjsuren D, Jadhav A, Maloney DJ, Simeonov A, Bohr VA, and Brosh RM Jr
- Subjects
- Biocatalysis, Cell Line, Tumor, Cell Proliferation drug effects, DNA metabolism, Enzyme Assays, Enzyme Inhibitors chemistry, Fluorometry, Humans, Inhibitory Concentration 50, Reproducibility of Results, Small Molecule Libraries chemistry, Werner Syndrome Helicase metabolism, Enzyme Inhibitors pharmacology, High-Throughput Screening Assays methods, Small Molecule Libraries analysis, Small Molecule Libraries pharmacology, Werner Syndrome Helicase antagonists & inhibitors
- Abstract
Werner syndrome (WS), an autosomal recessive genetic disorder, displays accelerated clinical symptoms of aging leading to a mean lifespan less than 50 years. The WS helicase-nuclease (WRN) is involved in many important pathways including DNA replication, recombination and repair. Replicating cells are dependent on helicase activity, leading to the pursuit of human helicases as potential therapeutic targets for cancer treatment. Small molecule inhibitors of DNA helicases can be used to induce synthetic lethality, which attempts to target helicase-dependent compensatory DNA repair pathways in tumor cells that are already genetically deficient in a specific pathway of DNA repair. Alternatively, helicase inhibitors may be useful as tools to study the specialized roles of helicases in replication and DNA repair. In this study, approximately 350,000 small molecules were screened based on their ability to inhibit duplex DNA unwinding by a catalytically active WRN helicase domain fragment in a high-throughput fluorometric assay to discover new non-covalent small molecule inhibitors of the WRN helicase. Select compounds were screened to exclude ones that inhibited DNA unwinding by other helicases in the screen, bound non-specifically to DNA, acted as irreversible inhibitors, or possessed unfavorable chemical properties. Several compounds were tested for their ability to impair proliferation of cultured tumor cells. We observed that two of the newly identified WRN helicase inhibitors inhibited proliferation of cancer cells in a lineage-dependent manner. These studies represent the first high-throughput screen for WRN helicase inhibitors and the results have implications for anti-cancer strategies targeting WRN in different cancer cells and genetic backgrounds., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
43. Enhanced mitochondrial DNA repair of the common disease-associated variant, Ser326Cys, of hOGG1 through small molecule intervention.
- Author
-
Baptiste BA, Katchur SR, Fivenson EM, Croteau DL, Rumsey WL, and Bohr VA
- Subjects
- A549 Cells, Animals, Cells, Cultured, DNA Damage, DNA Glycosylases genetics, Fibroblasts cytology, Fibroblasts drug effects, Fibroblasts metabolism, Herbicides adverse effects, Humans, Mice, Mice, Knockout, Mitochondria genetics, Mitochondria metabolism, Mitochondria pathology, Mutation, Oxidation-Reduction, Oxidative Stress drug effects, Paraquat adverse effects, Reactive Oxygen Species metabolism, Serine genetics, DNA Glycosylases metabolism, DNA Glycosylases physiology, DNA Repair, DNA, Mitochondrial genetics, Mitochondria drug effects, Serine metabolism, Small Molecule Libraries pharmacology
- Abstract
The common oxidatively generated lesion, 8-oxo-7,8-dihydroguanine (8-oxoGua), is removed from DNA by base excision repair. The glycosylase primarily charged with recognition and removal of this lesion is 8-oxoGuaDNA glycosylase 1 (OGG1). When left unrepaired, 8-oxodG alters transcription and is mutagenic. Individuals homozygous for the less active OGG1 allele, Ser326Cys, have increased risk of several cancers. Here, small molecule enhancers of OGG1 were identified and tested for their ability to stimulate DNA repair and protect cells from the environmental hazard paraquat (PQ). PQ-induced mtDNA damage was inversely proportional to the levels of OGG1 expression whereas stimulation of OGG1, in some cases, entirely abolished its cellular effects. The PQ-mediated decline of mitochondrial membrane potential or nuclear condensation were prevented by the OGG1 activators. In addition, in Ogg1
-/- mouse embryonic fibroblasts complemented with hOGG1S326C , there was increased cellular and mitochondrial reactive oxygen species compared to their wild type counterparts. Mitochondrial extracts from cells expressing hOGG1S326C were deficient in mitochondrial 8-oxodG incision activity, which was rescued by the OGG1 activators. These data demonstrate that small molecules can stimulate OGG1 activity with consequent cellular protection. Thus, OGG1-activating compounds may be useful in select humans to mitigate the deleterious effects of environmental oxidants and mutagens., (Copyright © 2018 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
44. NAD + supplementation normalizes key Alzheimer's features and DNA damage responses in a new AD mouse model with introduced DNA repair deficiency.
- Author
-
Hou Y, Lautrup S, Cordonnier S, Wang Y, Croteau DL, Zavala E, Zhang Y, Moritoh K, O'Connell JF, Baptiste BA, Stevnsner TV, Mattson MP, and Bohr VA
- Subjects
- Animals, Cognitive Dysfunction, DNA Damage, Gene Expression Regulation drug effects, Male, Mice, Mice, Transgenic, Neurogenesis drug effects, Niacinamide pharmacology, Pyridinium Compounds, Sirtuin 3 genetics, Sirtuin 3 metabolism, Sirtuins genetics, Sirtuins metabolism, tau Proteins metabolism, Alzheimer Disease, Disease Models, Animal, NAD pharmacology, Niacinamide analogs & derivatives
- Abstract
Emerging findings suggest that compromised cellular bioenergetics and DNA repair contribute to the pathogenesis of Alzheimer's disease (AD), but their role in disease-defining pathology is unclear. We developed a DNA repair-deficient 3xTgAD/Polβ
+/- mouse that exacerbates major features of human AD including phosphorylated Tau (pTau) pathologies, synaptic dysfunction, neuronal death, and cognitive impairment. Here we report that 3xTgAD/Polβ+/- mice have a reduced cerebral NAD+ /NADH ratio indicating impaired cerebral energy metabolism, which is normalized by nicotinamide riboside (NR) treatment. NR lessened pTau pathology in both 3xTgAD and 3xTgAD/Polβ+/- mice but had no impact on amyloid β peptide (Aβ) accumulation. NR-treated 3xTgAD/Polβ+/- mice exhibited reduced DNA damage, neuroinflammation, and apoptosis of hippocampal neurons and increased activity of SIRT3 in the brain. NR improved cognitive function in multiple behavioral tests and restored hippocampal synaptic plasticity in 3xTgAD mice and 3xTgAD/Polβ+/- mice. In general, the deficits between genotypes and the benefits of NR were greater in 3xTgAD/Polβ+/- mice than in 3xTgAD mice. Our findings suggest a pivotal role for cellular NAD+ depletion upstream of neuroinflammation, pTau, DNA damage, synaptic dysfunction, and neuronal degeneration in AD. Interventions that bolster neuronal NAD+ levels therefore have therapeutic potential for AD., Competing Interests: The authors declare no conflict of interest.- Published
- 2018
- Full Text
- View/download PDF
45. Cell cycle-dependent phosphorylation regulates RECQL4 pathway choice and ubiquitination in DNA double-strand break repair.
- Author
-
Lu H, Shamanna RA, de Freitas JK, Okur M, Khadka P, Kulikowicz T, Holland PP, Tian J, Croteau DL, Davis AJ, and Bohr VA
- Subjects
- 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, Cell Cycle, DNA Breaks, Double-Stranded, DNA End-Joining Repair, RecQ Helicases metabolism, Recombinational DNA Repair, Ubiquitination
- 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.
- Published
- 2017
- Full Text
- View/download PDF
46. NAD + in Aging: Molecular Mechanisms and Translational Implications.
- Author
-
Fang EF, Lautrup S, Hou Y, Demarest TG, Croteau DL, Mattson MP, and Bohr VA
- Subjects
- Alzheimer Disease pathology, Alzheimer Disease therapy, Animals, Cardiovascular Diseases pathology, Cardiovascular Diseases therapy, Humans, Muscular Atrophy pathology, Muscular Atrophy therapy, NAD, Parkinson Disease pathology, Parkinson Disease therapy, Aging metabolism, Alzheimer Disease metabolism, Cardiovascular Diseases metabolism, Energy Metabolism, Muscular Atrophy metabolism, Parkinson Disease metabolism
- Abstract
The coenzyme NAD
+ is critical in cellular bioenergetics and adaptive stress responses. Its depletion has emerged as a fundamental feature of aging that may predispose to a wide range of chronic diseases. Maintenance of NAD+ levels is important for cells with high energy demands and for proficient neuronal function. NAD+ depletion is detected in major neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases, cardiovascular disease and muscle atrophy. Emerging evidence suggests that NAD+ decrements occur in various tissues during aging, and that physiological and pharmacological interventions bolstering cellular NAD+ levels might retard aspects of aging and forestall some age-related diseases. Here, we discuss aspects of NAD+ biosynthesis, together with putative mechanisms of NAD+ action against aging, including recent preclinical and clinical trials., (Published by Elsevier Ltd.)- Published
- 2017
- Full Text
- View/download PDF
47. Recent Advances in Understanding Werner Syndrome.
- Author
-
Shamanna RA, Croteau DL, Lee JH, and Bohr VA
- Abstract
Aging, the universal phenomenon, affects human health and is the primary risk factor for major disease pathologies. Progeroid diseases, which mimic aging at an accelerated rate, have provided cues in understanding the hallmarks of aging. Mutations in DNA repair genes as well as in telomerase subunits are known to cause progeroid syndromes. Werner syndrome (WS), which is characterized by accelerated aging, is an autosomal-recessive genetic disorder. Hallmarks that define the aging process include genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulation of nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication. WS recapitulates these hallmarks of aging and shows increased incidence and early onset of specific cancers. Genome integrity and stability ensure the normal functioning of the cell and are mainly guarded by the DNA repair machinery and telomeres. WRN, being a RecQ helicase, protects genome stability by regulating DNA repair pathways and telomeres. Recent advances in WS research have elucidated WRN's role in DNA repair pathway choice regulation, telomere maintenance, resolution of complex DNA structures, epigenetic regulation, and stem cell maintenance., Competing Interests: Competing interests: The authors declare that they have no competing interests.No competing interests were disclosed.No competing interests were disclosed.No competing interests were disclosed.
- Published
- 2017
- Full Text
- View/download PDF
48. DNA Polymerase Beta Participates in Mitochondrial DNA Repair.
- Author
-
Sykora P, Kanno S, Akbari M, Kulikowicz T, Baptiste BA, Leandro GS, Lu H, Tian J, May A, Becker KA, Croteau DL, Wilson DM 3rd, Sobol RW, Yasui A, and Bohr VA
- Abstract
We have detected DNA polymerase beta (Polβ), known as a key nuclear base excision repair (BER) protein, in mitochondrial protein extracts derived from mammalian tissue and cells. Manipulation of the N-terminal sequence affected the amount of Polβ in the mitochondria. Using Polβ fragments, mitochondrion-specific protein partners were identified, with the interactors functioning mainly in DNA maintenance and mitochondrial import. Of particular interest was the identification of the proteins TWINKLE, SSBP1, and TFAM, all of which are mitochondrion-specific DNA effectors and are known to function in the nucleoid. Polβ directly interacted functionally with the mitochondrial helicase TWINKLE. Human kidney cells with Polβ knockout (KO) had higher endogenous mitochondrial DNA (mtDNA) damage. Mitochondrial extracts derived from heterozygous Polβ mouse tissue and KO cells had lower nucleotide incorporation activity. Mouse-derived Polβ null fibroblasts had severely affected metabolic parameters. Indeed, gene knockout of Polβ caused mitochondrial dysfunction, including reduced membrane potential and mitochondrial content. We show that Polβ is a mitochondrial polymerase involved in mtDNA maintenance and is required for mitochondrial homeostasis., (Copyright © 2017 American Society for Microbiology.)
- Published
- 2017
- Full Text
- View/download PDF
49. NAD + in DNA repair and mitochondrial maintenance.
- Author
-
Croteau DL, Fang EF, Nilsen H, and Bohr VA
- Subjects
- Animals, Cell Nucleus metabolism, Humans, Mice, Poly (ADP-Ribose) Polymerase-1 metabolism, Signal Transduction, Sirtuins metabolism, DNA Repair, Mitochondria metabolism, NAD metabolism
- Published
- 2017
- Full Text
- View/download PDF
50. Human RECQL1 participates in telomere maintenance.
- Author
-
Popuri V, Hsu J, Khadka P, Horvath K, Liu Y, Croteau DL, and Bohr VA
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.