Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Mol Ther Nucleic Acids ; 34: 102057, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37928442

ABSTRACT

Toxic gain-of-function mutations in superoxide dismutase 1 (SOD1) contribute to approximately 2%-3% of all amyotrophic lateral sclerosis (ALS) cases. Artificial microRNAs (amiRs) delivered by adeno-associated virus (AAV) have been proposed as a potential treatment option to silence SOD1 expression and mitigate disease progression. Primary microRNA (pri-miRNA) scaffolds are used in amiRs to shuttle a hairpin RNA into the endogenous miRNA pathway, but it is unclear whether different primary miRNA (pri-miRNA) scaffolds impact the potency and safety profile of the expressed amiR in vivo. In our process to develop an AAV amiR targeting SOD1, we performed a preclinical characterization of two pri-miRNA scaffolds, miR155 and miR30a, sharing the same guide strand sequence. We report that, while the miR155-based vector, compared with the miR30a-based vector, leads to a higher level of the amiR and more robust suppression of SOD1 in vitro and in vivo, it also presents significantly greater risks for CNS-related toxicities in vivo. Despite miR30a-based vector showing relatively lower potency, it can significantly delay the development of ALS-like phenotypes in SOD1-G93A mice and increase survival in a dose-dependent manner. These data highlight the importance of scaffold selection in the pursuit of highly efficacious and safe amiRs for RNA interference gene therapy.

2.
Gene Ther ; 30(5): 443-454, 2023 05.
Article in English | MEDLINE | ID: mdl-36450833

ABSTRACT

CRISPR-based gene editing technology represents a promising approach to deliver therapies for inherited disorders, including amyotrophic lateral sclerosis (ALS). Toxic gain-of-function superoxide dismutase 1 (SOD1) mutations are responsible for ~20% of familial ALS cases. Thus, current clinical strategies to treat SOD1-ALS are designed to lower SOD1 levels. Here, we utilized AAV-PHP.B variants to deliver CRISPR-Cas9 guide RNAs designed to disrupt the human SOD1 (huSOD1) transgene in SOD1G93A mice. A one-time intracerebroventricular injection of AAV.PHP.B-huSOD1-sgRNA into neonatal H11Cas9 SOD1G93A mice caused robust and sustained mutant huSOD1 protein reduction in the cortex and spinal cord, and restored motor function. Neonatal treatment also reduced spinal motor neuron loss, denervation at neuromuscular junction (NMJ) and muscle atrophy, diminished axonal damage and preserved compound muscle action potential throughout the lifespan of treated mice. SOD1G93A treated mice achieved significant disease-free survival, extending lifespan by more than 110 days. Importantly, a one-time intrathecal or intravenous injection of AAV.PHP.eB-huSOD1-sgRNA in adult H11Cas9 SOD1G93A mice, immediately before symptom onset, also extended lifespan by at least 170 days. We observed substantial protection against disease progression, demonstrating the utility of our CRISPR editing preclinical approach for target evaluation. Our approach uncovered key parameters (e.g., AAV capsid, Cas9 expression) that resulted in improved efficacy compared to similar approaches and can also serve to accelerate drug target validation.


Subject(s)
Amyotrophic Lateral Sclerosis , Mice , Humans , Animals , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/therapy , Superoxide Dismutase-1/genetics , Gene Editing , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Mice, Transgenic , Disease Models, Animal
3.
Sci Rep ; 12(1): 12044, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35835785

ABSTRACT

Successful weaning from ventilators not only improves the quality of life of patients, but also reduces medical expenses. The aim of this study was to explore the association between nutritional provision and successful ventilator weaning. In this retrospective study data from the Respiratory Care Center of Chung Shan Medical University Hospital between October, 2017 and July, 2019 on patient characteristics, amount of nutrition delivered, and clinical outcomes were retrieved. A total of 280 ventilated patients were enrolled and divided into successful extubation and failed weaning groups. There were 178 males (63.6%) and 102 females (36.4%) with a mean age of 67.3 ± 16.9 years. The successful extubation group consisted of patients who tended towards ideal body weight during the weaning process (BMI 23.9 ± 5.0 versus 22.7 ± 4.8 kg/m2, p < 0.001). Patients from both groups initially received the same nutritional intervention, while patients of successful extubation received significantly more calories and protein after weaning (23.8 ± 7.8 kcal versus 27.8 ± 9.1 kcal, p < 0.001 and 0.97 ± 0.36 g versus 1.14 ± 0.42 g, p < 0.001). Successful weaning was associated with higher survival rate (p = 0.016), shortened hospital stay (p = 0.001), and reduced medical costs (p < 0.001). Overall, nutritional support with high calories and protein was associated with the probability of successful ventilator weaning in patients undergoing prolonged mechanical ventilation. Adequate nutrition is a determinant of successful ventilator weaning.


Subject(s)
Quality of Life , Respiration, Artificial , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Nutritional Support , Retrospective Studies , Ventilator Weaning
4.
Hum Gene Ther ; 33(1-2): 103-108, 2022 01.
Article in English | MEDLINE | ID: mdl-34376063

ABSTRACT

Adeno-associated viruses (AAVs) are nonenveloped viruses that have become popular gene transfer vectors to deliver DNA to target cells in clinical gene therapy. Iodixanol-based density gradient is one of the widely used purification methods for serotype-independent AAVs. However, residual iodixanol in AAV could be a safety concern, and further purification to remove this process-related impurity is typically needed. An analytical assay with high sensitivity is essential for the detection of residual iodixanol to ensure the safety of AAV products. We developed a liquid chromatography-mass spectrometry method with the limit of quantification of 0.01 µg/mL for residual iodixanol measurement in AAVs. The method also demonstrated linearity over four orders of magnitude that allows quantifying a high iodixanol concentration in in-process samples with excellent recovery and accuracy. In addition, we further explored a highly efficient purification method for removal of the residual iodixanol, to minimize the safety concern from iodixanol as a process impurity.


Subject(s)
Dependovirus , Genetic Vectors , Chromatography, Liquid , Dependovirus/genetics , Genetic Therapy , Genetic Vectors/genetics , Mass Spectrometry , Triiodobenzoic Acids
5.
Mol Ther Methods Clin Dev ; 23: 68-77, 2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34589555

ABSTRACT

Robust assays to quantify adeno-associated virus (AAV) vector expression and potency are essential for gene therapy development. These assays inform the efficacy, safety, and pharmacodynamic profiles of AAV development candidates. Additionally, for gene downregulation strategies such as RNAi, knockdown of endogenous genes reflects the mechanism of action of such development candidates. Therefore, a method to quantify target mRNA repression is necessary for measuring vector potency both in vitro and in vivo. Here, we report the development of a one-step reverse-transcription droplet digital PCR (RT-ddPCR) method to analyze expression of AAV vectors and the potency of AAV-RNAi vectors. This one-step RT-ddPCR method simplifies the workflow, allows for duplexing reactions, and enables absolute quantification of transcripts without standard materials. With a gene augmentation vector, we demonstrate the application of RT-ddPCR in quantifying vector expression in vitro and in non-human primate (NHP) samples. This novel method is demonstrated to be precise and linear within the range of 0.05-25 ng of RNA input. Using an AAV-RNAi vector, we further demonstrate the utility of this RT-ddPCR method in quantifying potency. Orthogonal potency assays, including ELISA and functional readout, correlate well with RT-ddPCR results. Therefore, one-step RT-ddPCR can be implemented in the analytical and pharmacological characterization of AAV vectors.

7.
Gene Ther ; 28(10-11): 646-658, 2021 11.
Article in English | MEDLINE | ID: mdl-33558692

ABSTRACT

CRISPR-Cas systems have emerged as a powerful tool to generate genetic models for studying normal and diseased central nervous system (CNS). Targeted gene disruption at specific loci has been demonstrated successfully in non-dividing neurons. Despite its simplicity, high specificity and low cost, the efficiency of CRISPR-mediated knockout in vivo can be substantially impacted by many parameters. Here, we used CRISPR-Cas9 to disrupt the neuronal-specific gene, NeuN, and optimized key parameters to achieve effective gene knockout broadly in the CNS in postnatal mice. Three cell lines and two primary neuron cultures were used to validate the disruption of NeuN by single-guide RNAs (sgRNA) harboring distinct spacers and scaffold sequences. This triage identified an optimal sgRNA design with the highest NeuN disruption in in vitro and in vivo systems. To enhance CRISPR efficiency, AAV-PHP.B, a vector with superior neuronal transduction, was used to deliver this sgRNA in Cas9 mice via neonatal intracerebroventricular (ICV) injection. This approach resulted in 99.4% biallelic indels rate in the transduced cells, leading to greater than 70% reduction of total NeuN proteins in the cortex, hippocampus and spinal cord. This work contributes to the optimization of CRISPR-mediated knockout and will be beneficial for fundamental and preclinical research.


Subject(s)
CRISPR-Cas Systems , RNA, Guide, Kinetoplastida , Animals , Central Nervous System , Gene Editing/methods , Gene Knockout Techniques , Mice , Neurons/metabolism , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism
8.
Gene Ther ; 28(7-8): 456-468, 2021 08.
Article in English | MEDLINE | ID: mdl-33612827

ABSTRACT

Adeno-associated virus (AAV) transduction efficiency and tropism are conventionally determined by high expression of a fluorescent reporter gene. Emerging data has suggested that such conventional methods may underestimate AAV transduction for cells in which reporter expression from AAV vectors is undetectable. To explore an alternative method that captures AAV transduction in cells in which low expression of a cargo is sufficient for the intended activity, we sought after CRISPR/Cas9-mediated gene disruption. In this study, we use AAV to deliver CRISPR/guide RNA designed to abolish the genes NeuN, GFAP, or MOG expressed specifically in neurons, astrocytes, or oligodendrocytes respectively in the central nervous system (CNS) of mice. Abrogated expression of these cell-type-specific genes can be measured biochemically in CNS subregions and provides quantitative assessment of AAV transduction in these CNS cell types. By using this method, we compared CNS transduction of AAV9, AAV-PHP.B, and AAV-PHP.eB delivered via intracerebroventricular injection (ICV) in neonatal mice. We found both AAV-PHP.B and AAV-PHP.eB resulted in marked disruption of the NeuN gene by CRISPR/Cas9, significantly greater than AAV9 in several brain regions and spinal cord. In contrast, only modest disruption of the GFAP gene and the MOG gene was observed by all three AAV variants. Since the procedure of ICV circumvents the blood-brain barrier, our data suggests that, independent of their ability to cross the blood-brain barrier, AAV-PHP.B variants also exhibit remarkably improved neuronal transduction in the CNS. We anticipate this approach will facilitate profiling of AAV cellular tropism in murine CNS.


Subject(s)
Dependovirus , Genetic Vectors , Animals , CRISPR-Cas Systems , Central Nervous System , Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors/genetics , Mice , Neurons , Transduction, Genetic
9.
Sci Rep ; 6: 18335, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26783106

ABSTRACT

Impaired social interaction is a defining feature of autism spectrum disorder, a neurodevelopmental disorder that shows a strong male preponderance in prevalence. Studies have identified neural circuits, neuromodulators and genetic factors involved in social behaviors, but mechanistic understanding of gender-specific social deficits is lacking. We report that deletion of the caspase-3 gene, encoding a protease with functions in apoptosis and neural plasticity, alters specific social behaviors in male mice, while leaving females unaffected. Casp3(-/-) mice showed normal behavioral responses to olfactory cues from food, neutral chemical and biological sources. Both Casp3(-/-) males and females displayed robust social exploration, sociability, recognition and preference for an enclosed novel mouse in the three-chamber test. However, Casp3(-/-) males showed significantly reduced social interaction behaviors when exposed to a freely moving novel mouse, including decreased interaction time and diminished mounting. Thus caspase-3 is essential for a subset of social behaviors, but despite similar hyper-locomotion in both sexes, only male Casp3(-/-) mice exhibited social interaction deficits, which is interesting given the male bias of autism.


Subject(s)
Autism Spectrum Disorder/genetics , Behavior, Animal/physiology , Caspase 3/genetics , Locomotion/genetics , Animals , Autism Spectrum Disorder/physiopathology , Disease Models, Animal , Female , Interpersonal Relations , Male , Mice , Mice, Knockout
10.
J Neurosci ; 35(5): 2118-32, 2015 Feb 04.
Article in English | MEDLINE | ID: mdl-25653368

ABSTRACT

The ability to attend to relevant stimuli and to adapt dynamically as demands change is a core aspect of cognition, and one that is impaired in several neuropsychiatric diseases, including attention deficit/hyperactivity disorder. However, the cellular and molecular mechanisms underlying such cognitive adaptability are poorly understood. We found that deletion of the caspase-3 gene, encoding an apoptosis protease with newly discovered roles in neural plasticity, disrupts attention in mice while preserving multiple learning and memory capabilities. Attention-related deficits include distractibility, impulsivity, behavioral rigidity, and reduced habituation to novel stimuli. Excess exploratory activity in Casp3(-/-) mice was correlated with enhanced novelty-induced activity in the dentate gyrus, which may be related to our findings that caspase-3 is required for homeostatic synaptic plasticity in vitro and homeostatic expression of AMPA receptors in vivo in response to chronic or repeated stimuli. These results suggest an important role for caspase-3 in synaptic suppression of irrelevant stimuli.


Subject(s)
Attention Deficit Disorder with Hyperactivity/genetics , Attention , Caspase 3/deficiency , Homeostasis , Synapses/physiology , Animals , Attention Deficit Disorder with Hyperactivity/physiopathology , Caspase 3/genetics , Dentate Gyrus/metabolism , Dentate Gyrus/physiology , Gene Deletion , Maze Learning , Mice , Mice, Inbred C57BL , Neuronal Plasticity
11.
J Neurosci ; 33(14): 5924-9, 2013 Apr 03.
Article in English | MEDLINE | ID: mdl-23554474

ABSTRACT

Histone deacetylase 2 (HDAC2) negatively regulates excitatory synapse number and memory performance. However, whether HDAC2 regulation of excitatory synapses occurs in a cell-autonomous manner and whether HDAC2 regulates inhibitory synaptic functions are not well understood. To examine these aspects of HDAC2 function, we used sparse transfection of rat hippocampal slice cultures and whole-cell recordings in pyramidal neurons. HDAC2 knockdown (KD) in single postsynaptic pyramidal neurons enhanced, whereas HDAC2 overexpression (OE) reduced, excitatory synaptic transmission. Postsynaptic KD of HDAC2 also facilitated expression of long-term potentiation induced by subthreshold induction stimuli, without altering long-term depression. In contrast, HDAC2 KD reduced, whereas HDAC2 OE enhanced, inhibitory synaptic transmission. Alterations of postsynaptic GABA(A) receptors (GABA(A)Rs) likely underlie the impact of HDAC2 on inhibitory transmission. Consistent with this, we observed reduced transcript and protein levels of the GABA(A)R γ2 subunit and reduced surface expression of the α2 subunit after HDAC2 KD. Furthermore, we observed a reduction in synaptic but not tonic GABA(A)R currents by HDAC2 KD, suggesting that HDAC2 selectively affects synaptic abundance of functional GABA(A)Rs. Immunostaining for postsynaptic GABA(A)Rs confirmed that HDAC2 KD and OE can regulate the synaptic abundance of these receptors. Together, these results highlight a role for HDAC2 in suppressing synaptic excitation and enhancing synaptic inhibition of hippocampal neurons. Therefore, a shift in the balance of synaptic excitation versus inhibition favoring excitation could contribute to the beneficial effects of reducing HDAC2 function in wild-type mice or of inhibiting HDACs in models of cognitive impairment.


Subject(s)
CA1 Region, Hippocampal/cytology , Excitatory Postsynaptic Potentials/physiology , Histone Deacetylase 2/metabolism , Inhibitory Postsynaptic Potentials/physiology , Animals , Animals, Newborn , Cell Line, Transformed , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/genetics , Humans , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Male , Neurons , Neurotransmitter Agents/pharmacology , Patch-Clamp Techniques , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Transfection , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
12.
Nat Neurosci ; 14(5): 545-7, 2011 May.
Article in English | MEDLINE | ID: mdl-21441921
13.
Cell ; 141(5): 859-71, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20510932

ABSTRACT

NMDA receptor-dependent synaptic modifications, such as long-term potentiation (LTP) and long-term depression (LTD), are essential for brain development and function. LTD occurs mainly by the removal of AMPA receptors from the postsynaptic membrane, but the underlying molecular mechanisms remain unclear. Here, we show that activation of caspase-3 via mitochondria is required for LTD and AMPA receptor internalization in hippocampal neurons. LTD and AMPA receptor internalization are blocked by peptide inhibitors of caspase-3 and -9. In hippocampal slices from caspase-3 knockout mice, LTD is abolished whereas LTP remains normal. LTD is also prevented by overexpression of the anti-apoptotic proteins XIAP or Bcl-xL, and by a mutant Akt1 protein that is resistant to caspase-3 proteolysis. NMDA receptor stimulation that induces LTD transiently activates caspase-3 in dendrites, without causing cell death. These data indicate an unexpected causal link between the molecular mechanisms of apoptosis and LTD.


Subject(s)
Apoptosis , Caspase 3/metabolism , Hippocampus/metabolism , Long-Term Synaptic Depression , Receptors, AMPA/metabolism , Animals , Cells, Cultured , Cytochromes c/metabolism , Hippocampus/cytology , Long-Term Potentiation , Mice , Mice, Knockout , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , bcl-X Protein/metabolism
14.
Biotechnol Prog ; 26(4): 1001-7, 2010.
Article in English | MEDLINE | ID: mdl-20564357

ABSTRACT

Poly-gamma-glutamate (gamma-PGA) has applications in food, medical, cosmetic, animal feed, and wastewater industries. Bacillus subtilis DB430, which possesses the gamma-PGA synthesis ywsC-ywtAB genes in its chromosome, cannot produce gamma-PGA. An efficient synthetic expression control sequence (SECS) was introduced into the upstream region of the ywtABC genes, and this resulted in gamma-PGA-producing B. subtilis mutant strains. Mutant B. subtilis PGA6-2 stably produces high levels of gamma-PGA in medium A without supplementation of extra glutamic acid or ammonium chloride. The mutant B. subtilis PGA 6-2 is not only a gamma-PGA producer, but it is also a candidate for the genetic and metabolic engineering of gamma-PGA production.


Subject(s)
Bacillus subtilis/metabolism , Polyglutamic Acid/analogs & derivatives , Ammonium Chloride/pharmacology , Bacillus subtilis/drug effects , Bacillus subtilis/genetics , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Genetic Vectors/genetics , Glutamic Acid/pharmacology , Polyglutamic Acid/biosynthesis
15.
Exp Cell Res ; 314(8): 1789-803, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18387606

ABSTRACT

Eukaryote cells balance production of reactive oxygen species (ROS) with levels of anti-oxidant enzyme activity to maintain cellular redox homeostasis. Mitochondria are a major source of ROS, while many anti-oxidant genes are regulated by the Nrf2 transcription factor. Keap1, a redox-regulated substrate adaptor for a cullin-based ubiquitin ligase, targets Nrf2 for proteosome-mediated degradation and represses Nrf2-dependent gene expression. We have previously identified a member of the phosphoglycerate mutase family, PGAM5, as a Keap1-binding protein. In this report, we demonstrate that PGAM5 is targeted to the outer membrane of mitochondria by an N-terminal mitochondrial-localization sequence. Furthermore, we provide evidence that PGAM5 forms a ternary complex containing both Keap1 and Nrf2, in which the dimeric Keap1 protein simultaneously binds both PGAM5 and Nrf2 through their conserved E(S/T)GE motifs. Knockdown of either Keap1 or PGAM5 activates Nrf2-dependent gene expression. We suggest that this ternary complex provides a molecular framework for understanding how nuclear anti-oxidant gene expression is regulated in response to changes in mitochondrial function(s).


Subject(s)
Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Phosphoglycerate Mutase/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line , Cell Nucleus/genetics , Gene Expression Regulation , Humans , Kelch-Like ECH-Associated Protein 1 , Mitochondria/enzymology , Mitochondria/ultrastructure , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Molecular Sequence Data , Phosphoglycerate Mutase/chemistry , Phosphoglycerate Mutase/genetics , Phosphoprotein Phosphatases , Protein Isoforms/metabolism , Protein Sorting Signals
16.
Mol Cell Endocrinol ; 278(1-2): 52-62, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17920186

ABSTRACT

The orphan nuclear receptor, estrogen-related receptor beta (ERRbeta), shares a high degree of amino acid identity with estrogen receptor alpha (ERalpha). Although ERRbeta has been shown to be critical in embryo development, little is known about its functions and target genes. Here we report that the newly identified and most common human ortholog of ERRbeta--short-form hERRbeta (SFhERRbeta) potently represses the transcriptional activity of NF-E2 Related Factor 2 (Nrf2) on antioxidant response element (ARE)-mediated gene expression. Nrf2 is a main regulator of the expression of phase II detoxifying enzymes and antioxidant proteins in the cellular protection against oxidative stress. SFhERRbeta is the most potent inhibitor of Nrf2 transcriptional activity among the three ERR family members, ERRalpha, ERRbeta and ERRgamma. Additional analyses revealed that SFhERRbeta repressed Nrf2 activity likely through physical interaction in a complex with Nrf2, not by competing for the ARE DNA-binding sites, nor by decreasing Nrf2 protein concentration. By confocal immunofluorescence microscopy, SFhERRbeta alters the subcellular localization of Nrf2. Analyses using SFhERRbeta deletion mutants showed that SFhERRbeta interacts with Nrf2 through multiple sites. Our findings suggest that ERRbeta plays a novel functional role in the Nrf2-ARE pathway. By acting as a repressor of Nrf2, ERRbeta may be useful as a therapeutic target in cancer chemoprevention studies.


Subject(s)
Gene Expression Regulation , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , Receptors, Estrogen/metabolism , Repressor Proteins/metabolism , Animals , Antioxidants/pharmacology , COS Cells , Cell Line , Chlorocebus aethiops , Humans , NF-E2-Related Factor 2/analysis , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Structure, Tertiary , Receptors, Estrogen/genetics , Repressor Proteins/genetics , Response Elements/drug effects , Sequence Deletion , Transcription, Genetic/drug effects
17.
J Biol Chem ; 281(49): 37893-903, 2006 Dec 08.
Article in English | MEDLINE | ID: mdl-17046835

ABSTRACT

Keap1 is a BTB-Kelch substrate adaptor protein for a Cul3-dependent ubiquitin ligase complex that functions as a sensor for thiol-reactive chemopreventive compounds and oxidative stress. Inhibition of Keap1-dependent ubiquitination of the bZIP transcription factor Nrf2 enables Nrf2 to activate a cyto-protective transcriptional program that counters the damaging effects of oxidative stress. In this report we have identified a member of the phosphoglycerate mutase family, PGAM5, as a novel substrate for Keap1. The N terminus of the PGAM5 protein contains a conserved NXESGE motif that binds to the substrate binding pocket in the Kelch domain of Keap1, whereas the C-terminal PGAM domain binds Bcl-X(L). Keap1-dependent ubiquitination of PGAM5 results in proteasome-dependent degradation of PGAM5. Quinone-induced oxidative stress and the chemopreventive agent sulforaphane inhibit Keap1-dependent ubiquitination of PGAM5. The identification of PGAM5 as a novel substrate of Keap1 suggests that Keap1 regulates both transcriptional and post-transcriptional responses of mammalian cells to oxidative stress.


Subject(s)
Cell Cycle Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Autoantigens , Binding Sites , COS Cells , Cell Cycle Proteins/genetics , Cell Line , Chlorocebus aethiops , Humans , Intracellular Signaling Peptides and Proteins/genetics , Kelch-Like ECH-Associated Protein 1 , Membrane Proteins , Molecular Sequence Data , Oxidation-Reduction , Phylogeny , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Substrate Specificity , Transfection , Ubiquitin-Protein Ligases/genetics , bcl-X Protein/genetics , bcl-X Protein/metabolism
18.
EMBO J ; 25(15): 3605-17, 2006 Aug 09.
Article in English | MEDLINE | ID: mdl-16888629

ABSTRACT

Keap1 is a BTB-Kelch substrate adaptor protein that regulates steady-state levels of Nrf2, a bZIP transcription factor, in response to oxidative stress. We have determined the structure of the Kelch domain of Keap1 bound to a 16-mer peptide from Nrf2 containing a highly conserved DxETGE motif. The Nrf2 peptide contains two short antiparallel beta-strands connected by two overlapping type I beta-turns stabilized by the aspartate and threonine residues. The beta-turn region fits into a binding pocket on the top face of the Kelch domain and the glutamate residues form multiple hydrogen bonds with highly conserved residues in Keap1. Mutagenesis experiments confirmed the role of individual amino acids for binding of Nrf2 to Keap1 and for Keap1-mediated repression of Nrf2-dependent gene expression. Our results provide a detailed picture of how a BTB-Kelch substrate adaptor protein binds to its cognate substrate and will enable the rational design of novel chemopreventive agents.


Subject(s)
Intracellular Signaling Peptides and Proteins/chemistry , NF-E2-Related Factor 2/chemistry , Signal Transduction , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Cells, Cultured , Chlorocebus aethiops , Conserved Sequence , Dimerization , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kelch-Like ECH-Associated Protein 1 , Models, Molecular , Molecular Sequence Data , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Phosphorylation , Protein Binding , Protein Conformation , Protein Structure, Tertiary
19.
Mol Cell Biol ; 26(4): 1235-44, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16449638

ABSTRACT

The bZIP transcription factor Nrf2 controls a genetic program that protects cells from oxidative damage and maintains cellular redox homeostasis. Keap1, a BTB-Kelch protein, is the major upstream regulator of Nrf2. Keap1 functions as a substrate adaptor protein for a Cul3-dependent E3 ubiquitin ligase complex to repress steady-state levels of Nrf2 and Nrf2-dependent transcription. Cullin-dependent ubiquitin ligase complexes have been proposed to undergo dynamic cycles of assembly and disassembly that enable substrate adaptor exchange or recycling. In this report, we have characterized the importance of substrate adaptor recycling for regulation of Keap1-mediated repression of Nrf2. Association of Keap1 with Cul3 was decreased by ectopic expression of CAND1 and was increased by small interfering RNA (siRNA)-mediated knockdown of CAND1. However, both ectopic overexpression and siRNA-mediated knockdown of CAND1 decreased the ability of Keap1 to target Nrf2 for ubiquitin-dependent degradation, resulting in stabilization of Nrf2 and activation of Nrf2-dependent gene expression. Neddylation of Cul3 on Lys 712 is required for Keap1-dependent ubiquitination of Nrf2 in vivo. However, the K712R mutant Cul3 molecule, which is not neddylated, can still assemble with Keap1 into a functional ubiquitin ligase complex in vitro. These results provide support for a model in which substrate adaptor recycling is required for efficient substrate ubiquitination by cullin-dependent E3 ubiquitin ligase complexes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , NF-E2-Related Factor 2/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , CHO Cells , COS Cells , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Chlorocebus aethiops , Cricetinae , Cullin Proteins/genetics , Cullin Proteins/metabolism , HeLa Cells , Humans , NF-E2-Related Factor 2/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
20.
Neurochem Int ; 47(4): 298-307, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15955597

ABSTRACT

Nitric oxide (NO) release upon microglial cell activation has been implicated in the tissue injury and cell death in many neurodegenerative diseases. Recent studies have indicated the ability of interferon-gamma (IFNgamma) and lipopolysaccharides (LPS) to independently induce type II nitric oxide synthase (iNOS) expression and NO production in BV-2 microglial cells. However, a detailed comparison between the signaling pathways activating iNOS by these two agents has not been accomplished. Analysis of PKC isoforms revealed mainly the presence of PKCdelta, iota and lambda in BV-2 cells. Although both IFNgamma and LPS could specifically enhance the tyrosine phosphorylation of PKCdelta, treatment with IFNgamma induced a steady increase of phospho-PKCdelta for up to 1h, whereas treatment with LPS elevated phospho-PKCdelta levels only transiently, with peak activity at 5 min. Rottlerin, a specific inhibitor for PKCdelta, dose-dependently inhibited IFNgamma- and LPS-induced NO production. Despite the common involvement of PKCdelta, IFNgamma- but not LPS-induced NO production involved extracellular signal-regulated kinases (ERK1/2) cascade and IFNgamma-induced phosphorylation of ERK1/2 was mediated through PKC. On the other hand, LPS- but not IFNgamma-induced NO production was through stimulation of NF-kappaB activation and nuclear translocation to interact with DNA. These results demonstrated distinct signaling pathways for induction of iNOS by IFNgamma and LPS in BV-2 microglial cells.


Subject(s)
Interferon-gamma/physiology , Lipopolysaccharides/pharmacology , Microglia/immunology , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Signal Transduction/physiology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Animals , Cell Death/physiology , Cell Line, Transformed , Dose-Response Relationship, Drug , Encephalitis/metabolism , Encephalitis/physiopathology , Enzyme Inhibitors/pharmacology , Gliosis/metabolism , Gliosis/physiopathology , Interferon-gamma/pharmacology , Mice , Microglia/drug effects , Microglia/metabolism , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/drug effects , NF-kappa B/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase Type II , Phosphorylation/drug effects , Protein Kinase C/drug effects , Protein Kinase C/metabolism , Protein Kinase C-delta , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...