Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Proc Natl Acad Sci U S A ; 115(51): 13003-13008, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30509972

ABSTRACT

The instability of chromosome fragile sites is implicated as a causative factor in several human diseases, including cancer [for common fragile sites (CFSs)] and neurological disorders [for rare fragile sites (RFSs)]. Previous studies have indicated that problems arising during DNA replication are the underlying source of this instability. Although the role of replication stress in promoting instability at CFSs is well documented, much less is known about how the fragility of RFSs arises. Many RFSs, as exemplified by expansion of a CGG trinucleotide repeat sequence in the fragile X syndrome-associated FRAXA locus, exhibit fragility in response to folate deficiency or other forms of "folate stress." We hypothesized that such folate stress, through disturbing the replication program within the pathologically expanded repeats within FRAXA, would lead to mitotic abnormalities that exacerbate locus instability. Here, we show that folate stress leads to a dramatic increase in missegregation of FRAXA coupled with the formation of single-stranded DNA bridges in anaphase and micronuclei that contain the FRAXA locus. Moreover, chromosome X aneuploidy is seen when these cells are exposed to folate deficiency for an extended period. We propose that problematic FRAXA replication during interphase leads to a failure to disjoin the sister chromatids during anaphase. This generates further instability not only at FRAXA itself but also of chromosome X. These data have wider implications for the effects of folate deficiency on chromosome instability in human cells.


Subject(s)
Chromosome Fragile Sites , Chromosomes, Human, X , Folic Acid/metabolism , Fragile X Syndrome/pathology , Lymphocytes/pathology , Mitosis , Stress, Physiological , Cells, Cultured , DNA Replication , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/metabolism , Humans , Lymphocytes/metabolism , Male , Trinucleotide Repeat Expansion
2.
Hum Mol Genet ; 25(9): 1792-802, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26908614

ABSTRACT

Oxidative damage to mitochondria (MT) is a major mechanism for aging and neurodegeneration. We have developed a novel synthetic antioxidant, XJB-5-131, which directly targets MT, the primary site and primary target of oxidative damage. XJB-5-131 prevents the onset of motor decline in an HdhQ(150/150) mouse model for Huntington's disease (HD) if treatment starts early. Here, we report that XJB-5-131 attenuates or reverses disease progression if treatment occurs after disease onset. In animals with well-developed pathology, XJB-5-131 promotes weight gain, prevents neuronal death, reduces oxidative damage in neurons, suppresses the decline of motor performance or improves it, and reduces a graying phenotype in treated HdhQ(150/150) animals relative to matched littermate controls. XJB-5-131 holds promise as a clinical candidate for the treatment of HD.


Subject(s)
Cyclic N-Oxides/pharmacology , Disease Models, Animal , Huntington Disease/drug therapy , Mitochondria/drug effects , Motor Activity/drug effects , Oxidative Stress/drug effects , Animals , Behavior, Animal/drug effects , Cells, Cultured , Huntington Disease/metabolism , Huntington Disease/physiopathology , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/pathology , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Weight Loss/drug effects
3.
PLoS Genet ; 11(8): e1005267, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26247199

ABSTRACT

Huntington's Disease (HD) is caused by inheritance of a single disease-length allele harboring an expanded CAG repeat, which continues to expand in somatic tissues with age. The inherited disease allele expresses a toxic protein, and whether further somatic expansion adds to toxicity is unknown. We have created an HD mouse model that resolves the effects of the inherited and somatic expansions. We show here that suppressing somatic expansion substantially delays the onset of disease in littermates that inherit the same disease-length allele. Furthermore, a pharmacological inhibitor, XJB-5-131, inhibits the lengthening of the repeat tracks, and correlates with rescue of motor decline in these animals. The results provide evidence that pharmacological approaches to offset disease progression are possible.


Subject(s)
Cyclic N-Oxides/pharmacology , Huntington Disease/genetics , Trinucleotide Repeat Expansion/drug effects , Animals , Cyclic N-Oxides/therapeutic use , DNA Glycosylases/genetics , Disease Models, Animal , Disease Progression , Female , Huntington Disease/drug therapy , Huntington Disease/pathology , Male , Mice, Inbred C57BL , Mice, Knockout
4.
Hum Mol Genet ; 23(1): 129-44, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24021477

ABSTRACT

Loss of cholesterol homeostasis and altered vesicle trafficking have been detected in Huntington's disease (HD) cellular and animal models, yet the role of these dysfunctions in pathophysiology of HD is unknown. We demonstrate here that defects in caveolar-related cholesterol trafficking directly contribute to the mechanism of HD in vivo. We generated new mouse models that express mutant Huntington's protein (mhtt), but have partial or total loss of caveolin-1 (Cav1) expression. Fluorescence resonance energy transfer dequenching confirms a direct interaction between mhtt and Cav1. Mhtt-expressing neurons exhibited cholesterol accumulation and suppressed caveolar-related post-Golgi trafficking from endoplasmic reticulum/Golgi to plasma membrane. Loss or reduction of Cav1 expression in a knock-in HD mouse model rescues the cholesterol phenotype in neurons and significantly delays the onset of motor decline and development of neuronal inclusions. We propose that aberrant interaction between Cav1 and mhtt leads to altered cholesterol homeostasis and plays a direct causative role in the onset of HD pathophysiology in vivo.


Subject(s)
Caveolin 1/genetics , Caveolin 1/metabolism , Cholesterol/metabolism , Huntington Disease/genetics , Huntington Disease/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Animals , Cell Membrane/metabolism , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Fluorescence Resonance Energy Transfer , Gene Knock-In Techniques , HEK293 Cells , Humans , Huntingtin Protein , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Neurons/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phenotype
5.
Nat Rev Genet ; 11(11): 786-99, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20953213

ABSTRACT

Trinucleotide expansion underlies several human diseases. Expansion occurs during multiple stages of human development in different cell types, and is sensitive to the gender of the parent who transmits the repeats. Repair and replication models for expansions have been described, but we do not know whether the pathway involved is the same under all conditions and for all repeat tract lengths, which differ among diseases. Currently, researchers rely on bacteria, yeast and mice to study expansion, but these models differ substantially from humans. We need now to connect the dots among human genetics, pathway biochemistry and the appropriate model systems to understand the mechanism of expansion as it occurs in human disease.


Subject(s)
Chromosomal Instability/genetics , Growth and Development/genetics , Trinucleotide Repeats/genetics , Animals , Female , Human Development/physiology , Humans , Male , Mice , Models, Biological , Saccharomyces cerevisiae/genetics , Spermatogonia/metabolism , Trinucleotide Repeat Expansion/genetics , Trinucleotide Repeat Expansion/physiology
6.
Proc Natl Acad Sci U S A ; 108(42): E837-44, 2011 Oct 18.
Article in English | MEDLINE | ID: mdl-21960445

ABSTRACT

Insertion and deletion of small heteroduplex loops are common mutations in DNA, but why some loops are prone to mutation and others are efficiently repaired is unknown. Here we report that the mismatch recognition complex, MSH2/MSH3, discriminates between a repair-competent and a repair-resistant loop by sensing the conformational dynamics of their junctions. MSH2/MSH3 binds, bends, and dissociates from repair-competent loops to signal downstream repair. Repair-resistant Cytosine-Adenine-Guanine (CAG) loops adopt a unique DNA junction that traps nucleotide-bound MSH2/MSH3, and inhibits its dissociation from the DNA. We envision that junction dynamics is an active participant and a conformational regulator of repair signaling, and governs whether a loop is removed by MSH2/MSH3 or escapes to become a precursor for mutation.


Subject(s)
DNA Mismatch Repair , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA/chemistry , DNA/metabolism , MutS Homolog 2 Protein/chemistry , MutS Homolog 2 Protein/metabolism , Amino Acid Substitution , Base Pair Mismatch , Base Sequence , Binding Sites , DNA/genetics , DNA-Binding Proteins/genetics , Fluorescence Resonance Energy Transfer , Humans , In Vitro Techniques , Models, Molecular , Multiprotein Complexes , MutS Homolog 2 Protein/genetics , MutS Homolog 3 Protein , Mutagenesis, Site-Directed , Nucleic Acid Conformation , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
7.
J Biol Chem ; 287(4): 2328-41, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22102415

ABSTRACT

MRE11-RAD50 is a key early response protein for processing DNA ends of broken chromosomes for repair, yet how RAD50 nucleotide dynamics regulate MRE11 nuclease activity is poorly understood. We report here that ATP binding and ATP hydrolysis cause a striking butterfly-like opening and closing of the RAD50 subunits, and each structural state has a dramatic functional effect on MRE11. RAD50-MRE11 has an extended conformation in solution when MRE11 is an active nuclease. However, ATP binding to RAD50 induces a closed conformation, and in this state MRE11 is an endonuclease. ATP hydrolysis opens the RAD50-MRE11 complex, and MRE11 maintains exonuclease activity. Thus, ATP hydrolysis is a molecular switch that converts MRE11 from an endonuclease to an exonuclease. We propose a testable model in which the open-closed transitions are used by RAD50-MRE11 to discriminate among DNA ends and drive the choice of recombination pathways.


Subject(s)
Adenosine Triphosphate/metabolism , Archaeal Proteins/metabolism , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases/metabolism , Exodeoxyribonucleases/metabolism , Multienzyme Complexes/metabolism , Pyrococcus furiosus/enzymology , Recombination, Genetic/physiology , Adenosine Triphosphate/genetics , Archaeal Proteins/genetics , DNA, Archaeal/genetics , DNA, Archaeal/metabolism , DNA-Binding Proteins/genetics , Endodeoxyribonucleases/genetics , Exodeoxyribonucleases/genetics , Hydrolysis , Multienzyme Complexes/genetics , Protein Binding , Protein Structure, Quaternary , Pyrococcus furiosus/genetics
8.
Nature ; 447(7143): 447-52, 2007 May 24.
Article in English | MEDLINE | ID: mdl-17450122

ABSTRACT

Although oxidative damage has long been associated with ageing and neurological disease, mechanistic connections of oxidation to these phenotypes have remained elusive. Here we show that the age-dependent somatic mutation associated with Huntington's disease occurs in the process of removing oxidized base lesions, and is remarkably dependent on a single base excision repair enzyme, 7,8-dihydro-8-oxoguanine-DNA glycosylase (OGG1). Both in vivo and in vitro results support a 'toxic oxidation' model in which OGG1 initiates an escalating oxidation-excision cycle that leads to progressive age-dependent expansion. Age-dependent CAG expansion provides a direct molecular link between oxidative damage and toxicity in post-mitotic neurons through a DNA damage response, and error-prone repair of single-strand breaks.


Subject(s)
Aging/genetics , DNA Glycosylases/metabolism , Neurons/metabolism , Trinucleotide Repeat Expansion/genetics , Animals , Cell Line , DNA Breaks, Single-Stranded , DNA Damage , DNA Glycosylases/deficiency , DNA Glycosylases/genetics , DNA Repair/genetics , Female , Guanosine/analogs & derivatives , Guanosine/metabolism , Humans , Huntington Disease/genetics , Male , Mice , Models, Genetic , Oxidation-Reduction
9.
J Huntingtons Dis ; 11(2): 141-151, 2022.
Article in English | MEDLINE | ID: mdl-35404288

ABSTRACT

BACKGROUND: Mitochondria (MT) are energy "powerhouses" of the cell and the decline in their function from oxidative damage is strongly correlated in many diseases. To suppress oxygen damage, we have developed and applied XJB-5-131 as a targeted platform for neutralizing reactive oxygen species (ROS) directly in MT. Although the beneficial activity of XJB-5-131 is well documented, the mechanism of its protective effects is not yet fully understood. OBJECTIVE: Here, we elucidate the mechanism of protection for XJB-5-131, a mitochondrial targeted antioxidant and electron scavenger. METHODS: The Seahorse Flux Analyzer was used to probe the respiratory states of isolated mouse brain mitochondria treated with XJB-5-131 compared to controls. RESULTS: Surprisingly, there is no direct impact of XJB-5-131 radical scavenger on the electron flow through the electron transport chain. Rather, XJB-5-131 is a mild uncoupler of oxidative phosphorylation. The nitroxide moiety in XJB-5-131 acts as a superoxide dismutase mimic, which both extracts or donates electrons during redox reactions. The electron scavenging activity of XJB-5-131 prevents the leakage of electrons and reduces formation of superoxide anion, thereby reducing ROS. CONCLUSION: We show here that XJB-5-131 is a mild uncoupler of oxidative phosphorylation in MT. The mild uncoupling property of XJB-5-131 arises from its redox properties, which exert a protective effect by reducing ROS-induced damage without sacrificing energy production. Because mitochondrial decline is a common and central feature of toxicity, the favorable properties of XJB-5-131 are likely to be useful in treating Huntington's disease and a wide spectrum of neurodegenerative diseases for which oxidative damage is a key component. The mild uncoupling properties of XJB-5-131 suggest a valuable mechanism of action for the design of clinically effective antioxidants.


Subject(s)
Huntington Disease , Oxidative Phosphorylation , Animals , Cyclic N-Oxides/pharmacology , Mice , Oxidative Stress , Reactive Oxygen Species/pharmacology
10.
J Huntingtons Dis ; 11(1): 3-15, 2022.
Article in English | MEDLINE | ID: mdl-34924397

ABSTRACT

Due to large increases in the elderly populations across the world, age-related diseases are expected to expand dramatically in the coming years. Among these, neurodegenerative diseases will be among the most devastating in terms of their emotional and economic impact on patients, their families, and associated subsidized health costs. There is no currently available cure or rescue for dying brain cells. Viable therapeutics for any of these disorders would be a breakthrough and provide relief for the large number of affected patients and their families. Neurodegeneration is accompanied by elevated oxidative damage and inflammation. While natural antioxidants have largely failed in clinical trials, preclinical phenotyping of the unnatural, mitochondrial targeted nitroxide, XJB-5-131, bodes well for further translational development in advanced animal models or in humans. Here we consider the usefulness of synthetic antioxidants for the treatment of Huntington's disease. The mitochondrial targeting properties of XJB-5-131 have great promise. It is both an electron scavenger and an antioxidant, reducing both somatic expansion and toxicity simultaneously through the same redox mechanism. By quenching reactive oxygen species, XJB-5-131 breaks the cycle between the rise in oxidative damage during disease progression and the somatic growth of the CAG repeat which depends on oxidation.


Subject(s)
Huntington Disease , Aged , Animals , Antioxidants/therapeutic use , Cyclic N-Oxides/therapeutic use , Humans , Huntington Disease/drug therapy , Oxidative Stress
11.
Genes (Basel) ; 14(1)2022 12 29.
Article in English | MEDLINE | ID: mdl-36672839

ABSTRACT

Flap endonuclease 1 (FEN1) is an essential enzyme that removes RNA primers and base lesions during DNA lagging strand maturation and long-patch base excision repair (BER). It plays a crucial role in maintaining genome stability and integrity. FEN1 is also implicated in RNA processing and biogenesis. A recent study from our group has shown that FEN1 is involved in trinucleotide repeat deletion by processing the RNA strand in R-loops through BER, further suggesting that the enzyme can modulate genome stability by facilitating the resolution of R-loops. However, it remains unknown how FEN1 can process RNA to resolve an R-loop. In this study, we examined the FEN1 cleavage activity on the RNA:DNA hybrid intermediates generated during DNA lagging strand processing and BER in R-loops. We found that both human and yeast FEN1 efficiently cleaved an RNA flap in the intermediates using its endonuclease activity. We further demonstrated that FEN1 was recruited to R-loops in normal human fibroblasts and senataxin-deficient (AOA2) fibroblasts, and its R-loop recruitment was significantly increased by oxidative DNA damage. We showed that FEN1 specifically employed its endonucleolytic cleavage activity to remove the RNA strand in an R-loop during BER. We found that FEN1 coordinated its DNA and RNA endonucleolytic cleavage activity with the 3'-5' exonuclease of APE1 to resolve the R-loop. Our results further suggest that FEN1 employed its unique tracking mechanism to endonucleolytically cleave the RNA strand in an R-loop by coordinating with other BER enzymes and cofactors during BER. Our study provides the first evidence that FEN1 endonucleolytic cleavage can result in the resolution of R-loops via the BER pathway, thereby maintaining genome integrity.


Subject(s)
Flap Endonucleases , R-Loop Structures , Humans , DNA/genetics , DNA/metabolism , DNA Repair/genetics , Exonucleases/genetics , Flap Endonucleases/genetics , Flap Endonucleases/metabolism , Genomic Instability , RNA/genetics
13.
Sci Rep ; 11(1): 15598, 2021 08 02.
Article in English | MEDLINE | ID: mdl-34341363

ABSTRACT

Although some neurodegenerative diseases can be identified by behavioral characteristics relatively late in disease progression, we currently lack methods to predict who has developed disease before the onset of symptoms, when onset will occur, or the outcome of therapeutics. New biomarkers are needed. Here we describe spectral phenotyping, a new kind of biomarker that makes disease predictions based on chemical rather than biological endpoints in cells. Spectral phenotyping uses Fourier Transform Infrared (FTIR) spectromicroscopy to produce an absorbance signature as a rapid physiological indicator of disease state. FTIR spectromicroscopy has over the past been used in differential diagnoses of manifest disease. Here, we report that the unique FTIR chemical signature accurately predicts disease class in mouse with high probability in the absence of brain pathology. In human cells, the FTIR biomarker accurately predicts neurodegenerative disease class using fibroblasts as surrogate cells.


Subject(s)
Biomarkers/metabolism , Neurodegenerative Diseases/classification , Neurodegenerative Diseases/diagnosis , Spectroscopy, Fourier Transform Infrared , Animals , Animals, Newborn , Astrocytes/pathology , Cells, Cultured , Fibroblasts/pathology , Humans , Lipids/analysis , Mice, Inbred C57BL , Neurodegenerative Diseases/pathology , Phenotype , Reproducibility of Results
14.
J Biol Chem ; 284(41): 28352-28366, 2009 Oct 09.
Article in English | MEDLINE | ID: mdl-19674974

ABSTRACT

The oxidized DNA base 8-oxoguanine (8-oxoG) is implicated in neuronal CAG repeat expansion associated with Huntington disease, yet it is unclear how such a DNA base lesion and its repair might cause the expansion. Here, we discovered size-limited expansion of CAG repeats during repair of 8-oxoG in a wild-type mouse cell extract. This expansion was deficient in extracts from cells lacking pol beta and HMGB1. We demonstrate that expansion is mediated through pol beta multinucleotide gap-filling DNA synthesis during long-patch base excision repair. Unexpectedly, FEN1 promotes expansion by facilitating ligation of hairpins formed by strand slippage. This alternate role of FEN1 and the polymerase beta (pol beta) multinucleotide gap-filling synthesis is the result of uncoupling of the usual coordination between pol beta and FEN1. HMGB1 probably promotes expansion by stimulating APE1 and FEN1 in forming single strand breaks and ligatable nicks, respectively. This is the first report illustrating that disruption of pol beta and FEN1 coordination during long-patch BER results in CAG repeat expansion.


Subject(s)
DNA Polymerase beta/metabolism , Flap Endonucleases/metabolism , Trinucleotide Repeat Expansion , Animals , DNA/genetics , DNA/metabolism , DNA Damage , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA Polymerase beta/genetics , DNA Repair , Fibroblasts/cytology , Fibroblasts/physiology , Flap Endonucleases/genetics , Guanine/analogs & derivatives , Guanine/chemistry , Guanine/metabolism , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Humans , Mice , Mice, Knockout
15.
Nat Struct Mol Biol ; 12(8): 663-70, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16025128

ABSTRACT

Cells have evolved sophisticated DNA repair systems to correct damaged DNA. However, the human DNA mismatch repair protein Msh2-Msh3 is involved in the process of trinucleotide (CNG) DNA expansion rather than repair. Using purified protein and synthetic DNA substrates, we show that Msh2-Msh3 binds to CAG-hairpin DNA, a prime candidate for an expansion intermediate. CAG-hairpin binding inhibits the ATPase activity of Msh2-Msh3 and alters both nucleotide (ADP and ATP) affinity and binding interfaces between protein and DNA. These changes in Msh2-Msh3 function depend on the presence of A.A mispaired bases in the stem of the hairpin and on the hairpin DNA structure per se. These studies identify critical functional defects in the Msh2-Msh3-CAG hairpin complex that could misdirect the DNA repair process.


Subject(s)
Base Pair Mismatch/genetics , DNA Repair/genetics , DNA-Binding Proteins/metabolism , DNA/metabolism , Models, Genetic , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Trinucleotide Repeat Expansion/genetics , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Base Pairing , Base Sequence , DNA-Binding Proteins/genetics , Electrophoretic Mobility Shift Assay , Mice , Mice, Transgenic , Molecular Sequence Data , MutS Homolog 2 Protein , MutS Homolog 3 Protein , Protein Binding , Proteins/genetics , Proto-Oncogene Proteins/genetics
16.
DNA Repair (Amst) ; 7(7): 1121-34, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18472310

ABSTRACT

Mammalian cells have evolved sophisticated DNA repair systems to correct mispaired or damaged bases and extrahelical loops. Emerging evidence suggests that, in some cases, the normal DNA repair machinery is "hijacked" to become a causative factor in mutation and disease, rather than act as a safeguard of genomic integrity. In this review, we consider two cases in which active MMR leads to mutation or to cell death. There may be similar mechanisms by which uncoupling of normal MMR recognition from downstream repair allows triplet expansions underlying human neurodegenerative disease, or cell death in response to chemical lesion.


Subject(s)
DNA Mismatch Repair , Neurodegenerative Diseases/genetics , Trinucleotide Repeat Expansion , Animals , Cell Death , DNA Repair Enzymes , Humans , Mice , Models, Biological , Mutation , Neurodegenerative Diseases/metabolism
17.
BMC Neurosci ; 10: 73, 2009 Jul 08.
Article in English | MEDLINE | ID: mdl-19586540

ABSTRACT

BACKGROUND: Huntington's disease (HD) is a progressive neurodegenerative disorder caused by a CAG repeat expansion mutation in the coding region of a novel gene. The mechanism of HD is unknown. Most data suggest that polyglutamine-mediated aggregation associated with expression of mutant huntingtin protein (mhtt) contributes to the pathology. However, recent studies have identified early cellular dysfunctions that preclude aggregate formation. Suppression of aggregation is accepted as one of the markers of successful therapeutic approaches. Previously, we demonstrated that tricyclic pyrone (TP) compounds efficiently inhibited formation of amyloid-beta (Abeta) aggregates in cell and mouse models representing Alzheimer's Disease (AD). In the present study, we aimed to determine whether TP compounds could prevent aggregation and restore early cellular defects in primary embryonic striatal neurons from animal model representing HD. RESULTS: TP compounds effectively inhibit aggregation caused by mhtt in neurons and glial cells. Treatment with TP compounds also alleviated cholesterol accumulation and restored clathrin-independent endocytosis in HD neurons. CONCLUSION: We have found that TP compounds not only blocked mhtt-induced aggregation, but also alleviated early cellular dysfunctions that preclude aggregate formation. Our data suggest TP molecules may be used as lead compounds for prevention or treatment of multiple neurodegenerative diseases including HD and AD.


Subject(s)
Huntington Disease/pathology , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Pyrones/pharmacology , Animals , Cells, Cultured , Cholesterol/metabolism , Corpus Striatum/pathology , Endocytosis/drug effects , Huntingtin Protein , Huntington Disease/drug therapy , Huntington Disease/metabolism , Inclusion Bodies/drug effects , Mice , Mice, Transgenic , Pyrones/therapeutic use
18.
Toxicol Sci ; 172(2): 417-434, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31428778

ABSTRACT

Perfluorooctane sulfonate (PFOS) has been widely utilized in numerous industries. Due to long environmental and biological half-lives, PFOS is a major public health concern. Although the literature suggests that PFOS may induce neurotoxicity, neurotoxic mechanisms, and neuropathology are poorly understood. Thus, the primary goal of this study was to determine if PFOS is selectively neurotoxic and potentially relevant to specific neurological diseases. Nematodes (Caenorhabditis elegans) were exposed to PFOS or related per- and polyfluoroalkyl substances (PFAS) for 72 h and tested for evidence of neuropathology through examination of cholinergic, dopaminergic, gamma-amino butyric acid (GABA)ergic, and serotoninergic neuronal morphologies. Dopaminergic and cholinergic functional analyses were assessed through 1-nonanol and Aldicarb assay. Mechanistic studies assessed total reactive oxygen species, superoxide ions, and mitochondrial content. Finally, therapeutic approaches were utilized to further examine pathogenic mechanisms. Dopaminergic neuropathology occurred at lower exposure levels (25 ppm, approximately 50 µM) than required to produce neuropathology in GABAergic, serotonergic, and cholinergic neurons (100 ppm, approximately 200 µM). Further, PFOS exposure led to dopamine-dependent functional deficits, without altering acetylcholine-dependent paralysis. Mitochondrial content was affected by PFOS at far lower exposure level than required to induce pathology (≥1 ppm, approximately 2 µM). Perfluorooctane sulfonate exposure also enhanced oxidative stress. Further, mutation in mitochondrial superoxide dismutase rendered animals more vulnerable. Neuroprotective approaches such as antioxidants, PFAS-protein dissociation, and targeted (mitochondrial) radical and electron scavenging were neuroprotective, suggesting specific mechanisms of action. In general, other tested PFAS were less neurotoxic. The primary impact is to prompt research into potential adverse outcomes related to PFAS-induced dopaminergic neurotoxicity in humans.


Subject(s)
Alkanesulfonic Acids/toxicity , Caenorhabditis elegans/drug effects , Dopamine/metabolism , Environmental Pollutants/toxicity , Fluorocarbons/toxicity , Neurons/drug effects , Neurotoxicity Syndromes/metabolism , Alkanesulfonic Acids/metabolism , Animals , Antioxidants/pharmacology , Caenorhabditis elegans/metabolism , Cell Line , Environmental Pollutants/metabolism , Fluorocarbons/metabolism , Humans , Neurons/metabolism , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
19.
Cell Metab ; 29(6): 1258-1273.e11, 2019 06 04.
Article in English | MEDLINE | ID: mdl-30930170

ABSTRACT

The basis for region-specific neuronal toxicity in Huntington disease is unknown. Here, we show that region-specific neuronal vulnerability is a substrate-driven response in astrocytes. Glucose is low in HdhQ(150/150) animals, and astrocytes in each brain region adapt by metabolically reprogramming their mitochondria to use endogenous, non-glycolytic metabolites as an alternative fuel. Each region is characterized by distinct metabolic pools, and astrocytes adapt accordingly. The vulnerable striatum is enriched in fatty acids, and mitochondria reprogram by oxidizing them as an energy source but at the cost of escalating reactive oxygen species (ROS)-induced damage. The cerebellum is replete with amino acids, which are precursors for glucose regeneration through the pentose phosphate shunt or gluconeogenesis pathways. ROS is not elevated, and this region sustains little damage. While mhtt expression imposes disease stress throughout the brain, sensitivity or resistance arises from an adaptive stress response, which is inherently region specific. Metabolic reprogramming may have relevance to other diseases.


Subject(s)
Astrocytes/metabolism , Brain/pathology , Cellular Reprogramming/physiology , Huntingtin Protein/genetics , Huntington Disease/genetics , Metabolism/physiology , Neurons/pathology , Animals , Astrocytes/pathology , Brain/metabolism , Brain Mapping , Cells, Cultured , Disease Models, Animal , Disease Susceptibility/pathology , Disease Susceptibility/psychology , Glucose/metabolism , Huntington Disease/metabolism , Huntington Disease/pathology , Male , Metabolism/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Organ Specificity , Oxidation-Reduction , Reactive Oxygen Species/metabolism
20.
DNA Repair (Amst) ; 6(4): 517-29, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17129768

ABSTRACT

While a role for DNA glycosylase activity in base excision repair (BER) is well understood, there is mounting evidence to implicate cooperation of DNA glycosylases with components of repair pathways other than BER. The mechanisms by which DNA glycosylases interact with non-BER pathways are, in many cases, poorly understood. However, accumulating evidence indicates that crosstalk is common and may be as important in signaling repair as the canonical pathways themselves.


Subject(s)
DNA Damage , DNA Glycosylases/metabolism , DNA Mismatch Repair , DNA Repair Enzymes/metabolism , Animals , DNA Repair , Mice , Nucleotides/metabolism , Oxidation-Reduction , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL