64 results on '"Boudreau RL"'
Search Results
2. Resilient Modulus Test — Triaxial Cell Interaction
- Author
-
Boudreau, RL, primary and Wang, J, additional
- Full Text
- View/download PDF
3. Repeatability of the Resilient Modulus Test Procedure
- Author
-
Boudreau, RL, primary
- Full Text
- View/download PDF
4. Resilient Modulus — Pavement Subgrade Design Value
- Author
-
Boudreau, RL, primary
- Full Text
- View/download PDF
5. Applying Gene Silencing Technology to Contraception
- Author
-
Dissen, GA, primary, Lomniczi, A, additional, Boudreau, RL, additional, Chen, YH, additional, Davidson, BL, additional, and Ojeda, SR, additional
- Published
- 2012
- Full Text
- View/download PDF
6. Targeted Gene Silencing to Induce Permanent Sterility
- Author
-
Dissen, GA, primary, Lomniczi, A, additional, Boudreau, RL, additional, Chen, YH, additional, Davidson, BL, additional, and Ojeda, SR, additional
- Published
- 2012
- Full Text
- View/download PDF
7. Mitoregulin self-associates to form likely homo-oligomeric pore-like complexes.
- Author
-
Linzer CR, Stein CS, Witmer NH, Xu Z, Schnicker NJ, and Boudreau RL
- Abstract
We and others previously found that a misannotated long noncoding RNA encodes for a conserved mitochondrial transmembrane microprotein named Mitoregulin (Mtln). Beyond an established role for Mtln in lipid metabolism, Mtln has been shown to broadly influence mitochondria, boosting respiratory efficiency and Ca
2+ retention capacity, while lowering ROS, yet the underlying mechanisms remain unresolved. Prior studies have identified possible Mtln protein interaction partners; however, a lack of consensus persists, and no claims have been made about Mtln's structure. We noted two key published observations that seemingly remained overlooked: 1) endogenous Mtln co-immunoprecipitates with epitope-tagged Mtln at high efficiency, and 2) Mtln primarily appears to exist in a ∼66 kDa complex. To investigate if Mtln may self-oligomerize into higher-order complexes, we performed co-immunoprecipitation, computational modeling, and native gel assessments of Mtln-containing complexes in cells and tissues and tested whether synthetic Mtln protein itself forms oligomeric complexes. Our combined results provide strong support that Mtln self-associates and likely forms a hexameric pore-like structure., Competing Interests: The authors declare that no conflicts of interest exist., (© 2024 The Authors.)- Published
- 2024
- Full Text
- View/download PDF
8. MIRO1 controls energy production and proliferation of smooth muscle cells.
- Author
-
Qian L, Koval OM, Endoni BT, Juhr D, Stein CS, Allamargot C, Lin LH, Guo DF, Rahmouni K, Boudreau RL, Streeter J, Thiel WH, and Grumbach IM
- Abstract
Background: The outer mitochondrial Rho GTPase 1, MIRO1, mediates mitochondrial motility within cells, but implications for vascular smooth muscle cell (VSMC) physiology and its roles invascular diseases, such as neointima formation following vascular injury are widely unknown., Methods: An in vivo model of selective Miro1 deletion in VSMCs was generated, and the animals were subjected to carotid artery ligation. The molecular mechanisms relevant to VSMC proliferation were then explored in explanted VSMCs by imaging mitochondrial positioning and cristae structure and assessing the effects on ATP production, metabolic function and interactions with components of the electron transport chain (ETC)., Results: MIRO1 was robustly expressed in VSMCs within human atherosclerotic plaques and promoted VSMC proliferation and neointima formation in mice by blocking cell-cycle progression at G1/S, mitochondrial positioning, and PDGF-induced ATP production and respiration; overexpression of a MIRO1 mutant lacking the EF hands that are required for mitochondrial mobility did not fully rescue these effects. At the ultrastructural level, Miro1 deletion distorted the mitochondrial cristae and reduced the formation of super complexes and the activity of ETC complex I., Conclusions: Mitochondrial motility is essential for VSMC proliferation and relies on MIRO1. The EF-hands of MIRO1 regulate the intracellular positioning of mitochondria. Additionally, the absence of MIRO1 leads to distorted mitochondrial cristae and reduced ATP generation. Our findings demonstrate that motility is linked to mitochondrial ATP production. We elucidated two unrecognized mechanisms through which MIRO1 influences cell proliferation by modulating mitochondria: first, by managing mitochondrial placement via Ca
2+ -dependent EF hands, and second, by affecting cristae structure and ATP synthesis., Competing Interests: The authors have declared that no conflict of interest exists.- Published
- 2024
- Full Text
- View/download PDF
9. Upstream alternative polyadenylation in SCN5A produces a short transcript isoform encoding a mitochondria-localized NaV1.5 N-terminal fragment that influences cardiomyocyte respiration.
- Author
-
Witmer NH, McLendon JM, Stein CS, Yoon JY, Berezhnaya E, Elrod JW, London BL, and Boudreau RL
- Abstract
SCN5A encodes the cardiac voltage-gated Na+ channel, NaV1.5, that initiates action potentials. SCN5A gene variants cause arrhythmias and increased heart failure risk. Mechanisms controlling NaV1.5 expression and activity are not fully understood. We recently found a well-conserved alternative polyadenylation (APA) signal downstream of the first SCN5A coding exon. This yields a SCN5A-short transcript isoform expressed in several species (e.g. human, pig, and cat), though rodents lack this upstream APA. Reanalysis of transcriptome-wide cardiac APA-seq and mRNA-seq data shows reductions in both upstream APA usage and short/full-length SCN5A mRNA ratios in failing hearts. Knock-in of the human SCN5A APA sequence into mice is sufficient to enable expression of SCN5A -short transcript, while significantly decreasing expression of full-length SCN5A mRNA. Notably, SCN5A -short transcript encodes a novel protein (NaV1.5-NT), composed of an N-terminus identical to NaV1.5 and a unique C-terminus derived from intronic sequence. AAV9 constructs were able to achieve stable NaV1.5-NT expression in mouse hearts, and western blot of human heart tissues showed bands co-migrating with NaV1.5-NT transgene-derived bands. NaV1.5-NT is predicted to contain a mitochondrial targeting sequence and localizes to mitochondria in cultured cardiomyocytes and in mouse hearts. NaV1.5-NT expression in cardiomyocytes led to elevations in basal oxygen consumption rate, ATP production, and mitochondrial ROS, while depleting NADH supply. Native PAGE analyses of mitochondria lysates revealed that NaV1.5-NT expression resulted in increased levels of disassembled complex V subunits and accumulation of complex I-containing supercomplexes. Overall, we discovered that APA-mediated regulation of SCN5A produces a short transcript encoding NaV1.5-NT. Our data support that NaV1.5-NT plays a multifaceted role in influencing mitochondrial physiology: 1) by increasing basal respiration likely through promoting complex V conformations that enhance proton leak, and 2) by increasing overall respiratory efficiency and NADH consumption by enhancing formation and/or stability of complex I-containing respiratory supercomplexes, though the specific molecular mechanisms underlying each of these remain unresolved.
- Published
- 2024
- Full Text
- View/download PDF
10. microRNA-1 Regulates Metabolic Flexibility in Skeletal Muscle via Pyruvate Metabolism.
- Author
-
Ismaeel A, Peck BD, Montgomery MM, Burke BI, Goh J, Kang G, Franco AB, Xia Q, Goljanek-Whysall K, McDonagh B, McLendon JM, Koopmans PJ, Jacko D, Schaaf K, Bloch W, Gehlert S, Wen Y, Murach KA, Peterson CA, Boudreau RL, Fisher-Wellman KH, and McCarthy JJ
- Abstract
MicroRNA-1 (miR-1) is the most abundant miRNA in adult skeletal muscle. To determine the function of miR-1 in adult skeletal muscle, we generated an inducible, skeletal muscle-specific miR-1 knockout (KO) mouse. Integration of RNA-sequencing (RNA-seq) data from miR-1 KO muscle with Argonaute 2 enhanced crosslinking and immunoprecipitation sequencing (AGO2 eCLIP-seq) from human skeletal muscle identified miR-1 target genes involved with glycolysis and pyruvate metabolism. The loss of miR-1 in skeletal muscle induced cancer-like metabolic reprogramming, as shown by higher pyruvate kinase muscle isozyme M2 (PKM2) protein levels, which promoted glycolysis. Comprehensive bioenergetic and metabolic phenotyping combined with skeletal muscle proteomics and metabolomics further demonstrated that miR-1 KO induced metabolic inflexibility as a result of pyruvate oxidation resistance. While the genetic loss of miR-1 reduced endurance exercise performance in mice and in C. elegans, the physiological down-regulation of miR-1 expression in response to a hypertrophic stimulus in both humans and mice causes a similar metabolic reprogramming that supports muscle cell growth. Taken together, these data identify a novel post-translational mechanism of adult skeletal muscle metabolism regulation mediated by miR-1.
- Published
- 2024
- Full Text
- View/download PDF
11. Mitoregulin self-associates to form likely homo-oligomeric pore-like structures.
- Author
-
Linzer CR, Stein CS, Witmer NH, Xu Z, Schnicker NJ, and Boudreau RL
- Abstract
We and others previously found that a misannotated long noncoding RNA encodes for a conserved mitochondrial transmembrane microprotein named Mitoregulin (Mtln). Beyond an established role for Mtln in lipid metabolism, Mtln has also been shown to more broadly influence mitochondria, boosting respiratory efficiency and Ca
2+ retention capacity, while lowering ROS, yet the underlying mechanisms remain unresolved. Prior studies have identified possible Mtln protein interaction partners; however, a lack of consensus persists, and no claims have been made about Mtln's structure. We previously noted two key published observations that seemingly remained overlooked: 1) endogenous Mtln co-immunoprecipitates with epitope-tagged Mtln at high efficiency, and 2) Mtln primarily exists in a ∼66 kDa complex. To investigate if Mtln may self-oligomerize into higher-order complexes, we performed co-immunoprecipitation, protein modeling simulations, and native gel assessments of Mtln-containing complexes in cells and tissues, as well as tested whether synthetic Mtln protein itself forms oligomeric complexes. Our combined results provide strong support that Mtln self-associates and likely forms a hexameric pore-like structure.- Published
- 2024
- Full Text
- View/download PDF
12. Mitoregulin supports mitochondrial membrane integrity and protects against cardiac ischemia-reperfusion injury.
- Author
-
Stein CS, Zhang X, Witmer NH, Pennington ER, Shaikh SR, and Boudreau RL
- Abstract
We and others discovered a highly-conserved mitochondrial transmembrane microprotein, named Mitoregulin (Mtln), that supports lipid metabolism. We reported that Mtln strongly binds cardiolipin (CL), increases mitochondrial respiration and Ca
2+ retention capacities, and reduces reactive oxygen species (ROS). Here we extend our observation of Mtln-CL binding and examine Mtln influence on cristae structure and mitochondrial membrane integrity during stress. We demonstrate that mitochondria from constitutive- and inducible Mtln-knockout (KO) mice are susceptible to membrane freeze-damage and that this can be rescued by acute Mtln re-expression. In mitochondrial-simulated lipid monolayers, we show that synthetic Mtln decreases lipid packing and monolayer elasticity. Lipidomics revealed that Mtln-KO heart tissues show broad decreases in 22:6-containing lipids and increased cardiolipin damage/remodeling. Lastly, we demonstrate that Mtln-KO mice suffer worse myocardial ischemia-reperfusion injury, hinting at a translationally-relevant role for Mtln in cardioprotection. Our work supports a model in which Mtln binds cardiolipin and stabilizes mitochondrial membranes to broadly influence diverse mitochondrial functions, including lipid metabolism, while also protecting against stress.- Published
- 2024
- Full Text
- View/download PDF
13. Fndc5 is translated from an upstream ATG start codon and cleaved to produce irisin myokine precursor protein in humans and mice.
- Author
-
Witmer NH, Linzer CR, and Boudreau RL
- Subjects
- Humans, Animals, Mice, Protein Biosynthesis, Myokines, Fibronectins metabolism, Fibronectins genetics, Codon, Initiator genetics
- Abstract
Witmer et al. provide genomic and molecular evidence to demonstrate that Fndc5 (irisin myokine precursor protein) is translated in humans from an overlooked upstream ATG codon., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
14. A Specialized Centrosome-Proteasome Axis Mediates Proteostasis and Influences Cardiac Stress through Txlnb.
- Author
-
McLendon JM, Zhang X, Stein CS, Baehr LM, Bodine SC, and Boudreau RL
- Abstract
Background: Centrosomes localize to perinuclear foci where they serve multifunctional roles, arranging the microtubule organizing center (MTOC) and anchoring ubiquitin-proteasome system (UPS) machinery. In mature cardiomyocytes, centrosomal proteins redistribute into a specialized perinuclear cage-like structure, and a potential centrosome-UPS interface has not been studied. Taxilin-beta (Txlnb), a cardiomyocyte-enriched protein, belongs to a family of centrosome adapter proteins implicated in protein quality control. We hypothesize that Txlnb plays a key role in centrosomal-proteasomal crosstalk in cardiomyocytes., Methods: Integrative bioinformatics assessed centrosomal gene dysregulation in failing hearts. Txlnb gain/loss-of-function studies were conducted in cultured cardiomyocytes and mice. Txlnb's role in cardiac proteotoxicity and hypertrophy was examined using CryAB-R120G mice and transverse aortic constriction (TAC), respectively. Molecular modeling investigated Txlnb structure/function., Results: Human failing hearts show consistent dysregulation of many centrosome-associated genes, alongside UPS-related genes. Txlnb emerged as a candidate regulator of cardiomyocyte proteostasis that localizes to the perinuclear centrosomal compartment. Txlnb's interactome strongly supports its involvement in cytoskeletal, microtubule, and UPS processes, particularly centrosome-related functions. Overexpressing Txlnb in cardiomyocytes reduced ubiquitinated protein accumulation and enhanced proteasome activity during hypertrophy. Txlnb-knockout (KO) mouse hearts exhibit proteasomal insufficiency and altered cardiac growth, evidenced by ubiquitinated protein accumulation, decreased 26Sβ5 proteasome activity, and lower mass with age. In Cryab-R120G mice, Txlnb loss worsened heart failure, causing lower ejection fractions. After TAC, Txlnb-KO mice also showed reduced ejection fraction, increased heart mass, and elevated ubiquitinated protein accumulation. Investigations into the molecular mechanisms revealed that Txlnb-KO did not affect proteasomal subunit expression but led to the upregulation of Txlna and several centrosomal proteins (Cep63, Ofd1, and Tubg) suggesting altered centrosomal dynamics. Structural predictions support Txlnb's role as a specialized centrosomal-adapter protein bridging centrosomes with proteasomes, confirmed by microtubule-dependent perinuclear localization., Conclusions: Together, these data provide initial evidence connecting Txlnb to cardiac proteostasis, hinting at the potential importance of functional bridging between specialized centrosomes and UPS in cardiomyocytes.
- Published
- 2024
- Full Text
- View/download PDF
15. Modulation of miR-29 influences myocardial compliance likely through coordinated regulation of calcium handling and extracellular matrix.
- Author
-
Zhang X, McLendon JM, Peck BD, Chen B, Song LS, and Boudreau RL
- Abstract
MicroRNAs (miRNAs) control the expression of diverse subsets of target mRNAs, and studies have found miRNA dysregulation in failing hearts. Expression of miR-29 is abundant in heart, increases with aging, and is altered in cardiomyopathies. Prior studies demonstrate that miR-29 reduction via genetic knockout or pharmacologic blockade can blunt cardiac hypertrophy and fibrosis in mice. Surprisingly, this depended on specifically blunting miR-29 actions in cardiomyocytes versus fibroblasts. To begin developing more translationally relevant vectors, we generated a novel transgene-encoded miR-29 inhibitor (TuD-29) that can be incorporated into a viral-mediated gene therapy for cardioprotection. Here, we corroborate that miR-29 expression and activity is higher in cardiomyocytes versus fibroblasts and demonstrate that TuD-29 effectively blunts hypertrophic responses in cultured cardiomyocytes and mouse hearts. Furthermore, we found that adeno-associated virus (AAV)-mediated miR-29 overexpression in mouse hearts induces early diastolic dysfunction, whereas AAV:TuD-29 treatment improves cardiac output by increasing end-diastolic and stroke volumes. The integration of RNA sequencing and miRNA-target interactomes reveals that miR-29 regulates genes involved in calcium handling, cell stress and hypertrophy, metabolism, ion transport, and extracellular matrix remodeling. These investigations support a likely versatile role for miR-29 in influencing myocardial compliance and relaxation, potentially providing a unique therapeutic avenue to improve diastolic function in heart failure patients., Competing Interests: The authors declare no competing interests., (© 2023 The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
16. Correction: Escudero-Flórez et al. Dengue Virus Infection Alters Inter-Endothelial Junctions and Promotes Endothelial-Mesenchymal-Transition-like Changes in Human Microvascular Endothelial Cells. Viruses 2023, 15 , 1437.
- Author
-
Escudero-Flórez M, Torres-Hoyos D, Miranda-Brand Y, Boudreau RL, Gallego-Gómez JC, and Vicente-Manzanares M
- Abstract
Ryan L [...] .
- Published
- 2023
- Full Text
- View/download PDF
17. Dengue Virus Infection Alters Inter-Endothelial Junctions and Promotes Endothelial-Mesenchymal-Transition-Like Changes in Human Microvascular Endothelial Cells.
- Author
-
Escudero-Flórez M, Torres-Hoyos D, Miranda-Brand Y, Boudreau RL, Gallego-Gómez JC, and Vicente-Manzanares M
- Subjects
- Humans, Endothelial Cells, Culture Media, Conditioned pharmacology, Culture Media, Conditioned metabolism, Dengue Virus genetics, Dengue, Virus Diseases metabolism
- Abstract
Dengue virus (DENV) is a pathogenic arbovirus that causes human disease. The most severe stage of the disease (severe dengue) is characterized by vascular leakage, hypovolemic shock, and organ failure. Endothelial dysfunction underlies these phenomena, but the causal mechanisms of endothelial dysfunction are poorly characterized. This study investigated the role of c-ABL kinase in DENV-induced endothelial dysfunction. Silencing c-ABL with artificial miRNA or targeting its catalytic activity with imatinib revealed that c-ABL is required for the early steps of DENV infection. DENV-2 infection and conditioned media from DENV-infected cells increased endothelial expression of c-ABL and CRKII phosphorylation, promoted expression of mesenchymal markers, e.g., vimentin and N-cadherin, and decreased the levels of endothelial-specific proteins, e.g., VE-cadherin and ZO-1. These effects were reverted by silencing or inhibiting c-ABL. As part of the acquisition of a mesenchymal phenotype, DENV infection and treatment with conditioned media from DENV-infected cells increased endothelial cell motility in a c-ABL-dependent manner. In conclusion, DENV infection promotes a c-ABL-dependent endothelial phenotypic change that leads to the loss of intercellular junctions and acquisition of motility.
- Published
- 2023
- Full Text
- View/download PDF
18. Insulin and Insulin-Like Growth Factor 1 Signaling Preserves Sarcomere Integrity in the Adult Heart.
- Author
-
Riehle C, Weatherford ET, McCarty NS, Seei A, Jaishy BP, Manivel R, Galuppo P, Allamargot C, Hameed T, Boudreau RL, Bauersachs J, Weiss RM, and Abel ED
- Subjects
- Mice, Animals, Insulin Receptor Substrate Proteins metabolism, Insulin metabolism, Serum Response Factor metabolism, Sarcomeres metabolism, Myocytes, Cardiac metabolism, TOR Serine-Threonine Kinases metabolism, RNA, Messenger metabolism, Connexins metabolism, Insulin-Like Growth Factor I genetics, Heart Failure metabolism
- Abstract
Insulin and insulin-like growth factor 1 (IGF1) signaling is transduced by insulin receptor substrate 1 (IRS1) and IRS2. To elucidate physiological and redundant roles of insulin and IGF1 signaling in adult hearts, we generated mice with inducible cardiomyocyte-specific deletion of insulin and IGF1 receptors or IRS1 and IRS2. Both models developed dilated cardiomyopathy, and most mice died by 8 weeks post-gene deletion. Heart failure was characterized by cardiomyocyte loss and disarray, increased proapoptotic signaling, and increased autophagy. Suppression of autophagy by activating mTOR signaling did not prevent heart failure. Transcriptional profiling revealed reduced serum response factor (SRF) transcriptional activity and decreased mRNA levels of genes encoding sarcomere and gap junction proteins as early as 3 days post-gene deletion, in concert with ultrastructural evidence of sarcomere disruption and intercalated discs within 1 week after gene deletion. These data confirm conserved roles for constitutive insulin and IGF1 signaling in suppressing autophagic and apoptotic signaling in the adult heart. The present study also identifies an unexpected role for insulin and IGF1 signaling in regulating an SRF-mediated transcriptional program, which maintains expression of genes encoding proteins that support sarcomere integrity in the adult heart, reduction of which results in rapid development of heart failure.
- Published
- 2022
- Full Text
- View/download PDF
19. Knockout of Sorbin And SH3 Domain Containing 2 (Sorbs2) in Cardiomyocytes Leads to Dilated Cardiomyopathy in Mice.
- Author
-
McLendon JM, Zhang X, Matasic DS, Kumar M, Koval OM, Grumbach IM, Sadayappan S, London B, and Boudreau RL
- Subjects
- Adult, Animals, Disease Models, Animal, Humans, Infant, Mice, Mice, Knockout, Myocytes, Cardiac metabolism, RNA-Binding Proteins genetics, src Homology Domains, Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Cardiomyopathy, Dilated, Heart Failure, RNA-Binding Proteins metabolism
- Abstract
Background Sorbin and SH3 domain containing 2 (Sorbs2) protein is a cytoskeletal adaptor with an emerging role in cardiac biology and disease; yet, its potential relevance to adult-onset cardiomyopathies remains underexplored. Sorbs2 global knockout mice display lethal arrhythmogenic cardiomyopathy; however, the causative mechanisms remain unclear. Herein, we examine Sorbs2 dysregulation in heart failure, characterize novel Sorbs2 cardiomyocyte-specific knockout mice (Sorbs2-cKO), and explore associations between Sorbs2 genetic variations and human cardiovascular disease. Methods and Results Bioinformatic analyses show myocardial Sorbs2 mRNA is consistently upregulated in humans with adult-onset cardiomyopathies and in heart failure models. We generated Sorbs2-cKO mice and report that they develop progressive systolic dysfunction and enlarged cardiac chambers, and they die with congestive heart failure at about 1 year old. After 3 months, Sorbs2-cKO mice begin to show atrial enlargement and P-wave anomalies, without dysregulation of action potential-associated ion channel and gap junction protein expressions. After 6 months, Sorbs2-cKO mice exhibit impaired contractility in dobutamine-treated hearts and skinned myofibers, without dysregulation of contractile protein expressions. From our comprehensive survey of potential mechanisms, we found that within 4 months, Sorbs2-cKO hearts have defective microtubule polymerization and compensatory upregulation of structural cytoskeletal and adapter proteins, suggesting that this early intracellular structural remodeling is responsible for contractile dysfunction. Finally, we identified genetic variants that associate with decreased Sorbs2 expression and human cardiac phenotypes, including conduction abnormalities, atrial enlargement, and dilated cardiomyopathy, consistent with Sorbs2-cKO mice phenotypes. Conclusions Our studies show that Sorbs2 is essential for maintaining structural integrity in cardiomyocytes, likely through strengthening the interactions between microtubules and other cytoskeletal proteins at cross-link sites.
- Published
- 2022
- Full Text
- View/download PDF
20. Modulation of miR-181 influences dopaminergic neuronal degeneration in a mouse model of Parkinson's disease.
- Author
-
Stein CS, McLendon JM, Witmer NH, and Boudreau RL
- Abstract
Parkinson's disease (PD) is caused by the loss of dopaminergic (DA) neurons in the substantia nigra (SN). Although PD pathogenesis is not fully understood, studies implicate perturbations in gene regulation, mitochondrial function, and neuronal activity. MicroRNAs (miRs) are small gene regulatory RNAs that inhibit diverse subsets of target mRNAs, and several studies have noted miR expression alterations in PD brains. For example, miR-181a is abundant in the brain and is increased in PD patient brain samples; however, the disease relevance of this remains unclear. Here, we show that miR-181 target mRNAs are broadly downregulated in aging and PD brains. To address whether the miR-181 family plays a role in PD pathogenesis, we generated adeno-associated viruses (AAVs) to overexpress and inhibit the miR-181 isoforms. After co-injection with AAV overexpressing alpha-synuclein (aSyn) into mouse SN (PD model), we found that moderate miR-181a/b overexpression exacerbated aSyn-induced DA neuronal loss, whereas miR-181 inhibition was neuroprotective relative to controls (GFP alone and/or scrambled RNA). Also, prolonged miR-181 overexpression in SN alone elicited measurable neurotoxicity that is coincident with an increased immune response. mRNA-seq analyses revealed that miR-181a/b inhibits genes involved in synaptic transmission, neurite outgrowth, and mitochondrial respiration, along with several genes having known protective roles and genetic links in PD., Competing Interests: The authors declare no competing interests., (© 2022 The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
21. Human miRNA miR-675 inhibits DUX4 expression and may be exploited as a potential treatment for Facioscapulohumeral muscular dystrophy.
- Author
-
Saad NY, Al-Kharsan M, Garwick-Coppens SE, Chermahini GA, Harper MA, Palo A, Boudreau RL, and Harper SQ
- Subjects
- Adult, Aged, Animals, Cell Death drug effects, Disease Models, Animal, Drug Delivery Systems, Female, Genetic Therapy, HEK293 Cells, Humans, Mice, Mice, Inbred C57BL, MicroRNAs metabolism, Muscle, Skeletal metabolism, Muscle, Skeletal pathology, Muscular Diseases, Muscular Dystrophy, Facioscapulohumeral pathology, Open Reading Frames drug effects, RNA Interference, Gene Expression Regulation drug effects, Homeodomain Proteins drug effects, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, MicroRNAs genetics, MicroRNAs pharmacology, Muscular Dystrophy, Facioscapulohumeral drug therapy
- Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is a potentially devastating myopathy caused by de-repression of the DUX4 gene in skeletal muscles. Effective therapies will likely involve DUX4 inhibition. RNA interference (RNAi) is one powerful approach to inhibit DUX4, and we previously described a RNAi gene therapy to achieve DUX4 silencing in FSHD cells and mice using engineered microRNAs. Here we report a strategy to direct RNAi against DUX4 using the natural microRNA miR-675, which is derived from the lncRNA H19. Human miR-675 inhibits DUX4 expression and associated outcomes in FSHD cell models. In addition, miR-675 delivery using gene therapy protects muscles from DUX4-associated death in mice. Finally, we show that three known miR-675-upregulating small molecules inhibit DUX4 and DUX4-activated FSHD biomarkers in FSHD patient-derived myotubes. To our knowledge, this is the first study demonstrating the use of small molecules to suppress a dominant disease gene using an RNAi mechanism., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
22. Modulation of the cardiac sodium channel Na V 1.5 peak and late currents by NAD + precursors.
- Author
-
Matasic DS, Yoon JY, McLendon JM, Mehdi H, Schmidt MS, Greiner AM, Quinones P, Morgan GM, Boudreau RL, Irani K, Brenner C, and London B
- Subjects
- Acetylation drug effects, Animals, Dietary Supplements, HEK293 Cells, Humans, Lysine metabolism, Metabolome, Mice, Inbred C57BL, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Niacinamide analogs & derivatives, Niacinamide chemistry, Niacinamide pharmacology, Phosphorylation drug effects, Pyridinium Compounds chemistry, Pyridinium Compounds pharmacology, Rats, Sprague-Dawley, Ion Channel Gating drug effects, Myocardium metabolism, NAD metabolism, NAV1.5 Voltage-Gated Sodium Channel metabolism
- Abstract
Rationale: The cardiac sodium channel Na
V 1.5, encoded by SCN5A, produces the rapidly inactivating depolarizing current INa that is responsible for the initiation and propagation of the cardiac action potential. Acquired and inherited dysfunction of NaV 1.5 results in either decreased peak INa or increased residual late INa (INa,L ), leading to tachy/bradyarrhythmias and sudden cardiac death. Previous studies have shown that increased cellular NAD+ and NAD+ /NADH ratio increase INa through suppression of mitochondrial reactive oxygen species and PKC-mediated NaV 1.5 phosphorylation. In addition, NAD+ -dependent deacetylation of NaV 1.5 at K1479 by Sirtuin 1 increases NaV 1.5 membrane trafficking and INa . The role of NAD+ precursors in modulating INa remains unknown., Objective: To determine whether and by which mechanisms the NAD+ precursors nicotinamide riboside (NR) and nicotinamide (NAM) affect peak INa and INa,L in vitro and cardiac electrophysiology in vivo., Methods and Results: The effects of NAD+ precursors on the NAD+ metabolome and electrophysiology were studied using HEK293 cells expressing wild-type and mutant NaV 1.5, rat neonatal cardiomyocytes (RNCMs), and mice. NR increased INa in HEK293 cells expressing NaV 1.5 (500 μM: 51 ± 18%, p = .02, 5 mM: 59 ± 22%, p = .03) and RNCMs (500 μM: 60 ± 26%, p = .02, 5 mM: 74 ± 39%, p = .03) while reducing INa,L at the higher concentration (RNCMs, 5 mM: -45 ± 11%, p = .04). NR (5 mM) decreased NaV 1.5 K1479 acetylation but increased INa in HEK293 cells expressing a mutant form of NaV 1.5 with disruption of the acetylation site (NaV 1.5-K1479A). Disruption of the PKC phosphorylation site abolished the effect of NR on INa . Furthermore, NAM (5 mM) had no effect on INa in RNCMs or in HEK293 cells expressing wild-type NaV 1.5, but increased INa in HEK293 cells expressing NaV 1.5-K1479A. Dietary supplementation with NR for 10-12 weeks decreased QTc in C57BL/6 J mice (0.35% NR: -4.9 ± 2.0%, p = .14; 1.0% NR: -9.5 ± 2.8%, p = .01)., Conclusions: NAD+ precursors differentially regulate NaV 1.5 via multiple mechanisms. NR increases INa , decreases INa,L , and warrants further investigation as a potential therapy for arrhythmic disorders caused by NaV 1.5 deficiency and/or dysfunction., Competing Interests: Declaration of Competing Interest DSM: None, JY: None, JMM: None, HM: None, AMG: None, MSS: None, PQ: None, GMM: None, RLB: None, KI: None, CB: a stockholder and member of the scientific advisory board of ChromaDex and a co-founder of ProHealthspan, which manufacture, distribute and sell nicotinamide riboside as a nutritional supplement, BL: None., (Copyright © 2020 Elsevier Ltd. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
23. Targeting Calpain for Heart Failure Therapy: Implications From Multiple Murine Models.
- Author
-
Wang Y, Chen B, Huang CK, Guo A, Wu J, Zhang X, Chen R, Chen C, Kutschke W, Weiss RM, Boudreau RL, Margulies KB, Hong J, and Song LS
- Abstract
Heart failure remains a major cause of morbidity and mortality in developed countries. There is still a strong need to devise new mechanism-based treatments for heart failure. Numerous studies have suggested the importance of the Ca
2+ -dependent protease calpain in cardiac physiology and pathology. However, no drugs are currently under development or testing in human patients to target calpain for heart failure treatment. Herein the data demonstrate that inhibition of calpain activity protects against deleterious ultrastructural remodeling and cardiac dysfunction in multiple rodent models of heart failure, providing compelling evidence that calpain inhibition is a promising therapeutic strategy for heart failure treatment.- Published
- 2018
- Full Text
- View/download PDF
24. When Knowing "Enough" May Still Not Be Enough.
- Author
-
Matkovich SJ and Boudreau RL
- Subjects
- Animals, Gene Regulatory Networks, Humans, MicroRNAs metabolism, RNA Processing, Post-Transcriptional, Transcriptome, Heart Diseases genetics, MicroRNAs genetics, Myocardium metabolism
- Published
- 2018
- Full Text
- View/download PDF
25. Mitoregulin: A lncRNA-Encoded Microprotein that Supports Mitochondrial Supercomplexes and Respiratory Efficiency.
- Author
-
Stein CS, Jadiya P, Zhang X, McLendon JM, Abouassaly GM, Witmer NH, Anderson EJ, Elrod JW, and Boudreau RL
- Subjects
- Amino Acid Sequence, Animals, Calcium metabolism, Cardiolipins chemistry, Cardiolipins metabolism, Electron Transport Chain Complex Proteins chemistry, Electron Transport Chain Complex Proteins metabolism, Fatty Acids chemistry, Fatty Acids metabolism, HeLa Cells, Humans, Mice, Mice, Knockout, Mitochondrial Membranes metabolism, Mitochondrial Proteins antagonists & inhibitors, Mitochondrial Proteins genetics, Oxidation-Reduction, Protein Binding, RNA Interference, RNA, Small Interfering metabolism, Reactive Oxygen Species metabolism, Sequence Alignment, Mitochondria metabolism, Mitochondrial Proteins metabolism, RNA, Long Noncoding metabolism
- Abstract
Mitochondria are composed of many small proteins that control protein synthesis, complex assembly, metabolism, and ion and reactive oxygen species (ROS) handling. We show that a skeletal muscle- and heart-enriched long non-coding RNA, LINC00116, encodes a highly conserved 56-amino-acid microprotein that we named mitoregulin (Mtln). Mtln localizes to the inner mitochondrial membrane, where it binds cardiolipin and influences protein complex assembly. In cultured cells, Mtln overexpression increases mitochondrial membrane potential, respiration rates, and Ca
2+ retention capacity while decreasing mitochondrial ROS and matrix-free Ca2+ . Mtln-knockout mice display perturbations in mitochondrial respiratory (super)complex formation and activity, fatty acid oxidation, tricarboxylic acid (TCA) cycle enzymes, and Ca2+ retention capacity. Blue-native gel electrophoresis revealed that Mtln co-migrates alongside several complexes, including the complex I assembly module, complex V, and supercomplexes. Under denaturing conditions, Mtln remains in high-molecular-weight complexes, supporting its role as a sticky molecular tether that enhances respiratory efficiency by bolstering protein complex assembly and/or stability., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
26. A common variant alters SCN5A-miR-24 interaction and associates with heart failure mortality.
- Author
-
Zhang X, Yoon JY, Morley M, McLendon JM, Mapuskar KA, Gutmann R, Mehdi H, Bloom HL, Dudley SC, Ellinor PT, Shalaby AA, Weiss R, Tang WHW, Moravec CS, Singh M, Taylor AL, Yancy CW, Feldman AM, McNamara DM, Irani K, Spitz DR, Breheny P, Margulies KB, London B, and Boudreau RL
- Subjects
- Action Potentials, Aged, Alleles, Animals, Binding Sites, Death, Sudden, Cardiac, Female, Gene Expression Profiling, Genotype, Heart Conduction System physiopathology, Heart Rate, Homozygote, Humans, Linkage Disequilibrium, Male, Mice, Middle Aged, Oligonucleotide Array Sequence Analysis, Patch-Clamp Techniques, Polymorphism, Single Nucleotide, Rats, Sprague-Dawley, Heart Failure genetics, MicroRNAs genetics, NAV1.5 Voltage-Gated Sodium Channel genetics
- Abstract
SCN5A encodes the voltage-gated Na+ channel NaV1.5 that is responsible for depolarization of the cardiac action potential and rapid intercellular conduction. Mutations disrupting the SCN5A coding sequence cause inherited arrhythmias and cardiomyopathy, and single-nucleotide polymorphisms (SNPs) linked to SCN5A splicing, localization, and function associate with heart failure-related sudden cardiac death. However, the clinical relevance of SNPs that modulate SCN5A expression levels remains understudied. We recently generated a transcriptome-wide map of microRNA (miR) binding sites in human heart, evaluated their overlap with common SNPs, and identified a synonymous SNP (rs1805126) adjacent to a miR-24 site within the SCN5A coding sequence. This SNP was previously shown to reproducibly associate with cardiac electrophysiological parameters, but was not considered to be causal. Here, we show that miR-24 potently suppresses SCN5A expression and that rs1805126 modulates this regulation. We found that the rs1805126 minor allele associates with decreased cardiac SCN5A expression and that heart failure subjects homozygous for the minor allele have decreased ejection fraction and increased mortality, but not increased ventricular tachyarrhythmias. In mice, we identified a potential basis for this in discovering that decreased Scn5a expression leads to accumulation of myocardial reactive oxygen species. Together, these data reiterate the importance of considering the mechanistic significance of synonymous SNPs as they relate to miRs and disease, and highlight a surprising link between SCN5A expression and nonarrhythmic death in heart failure.
- Published
- 2018
- Full Text
- View/download PDF
27. The Next Best Thing in Cardiovascular Research: Highlights From the Basic Cardiovascular Sciences Scientific Sessions.
- Author
-
Boudreau RL
- Subjects
- Diffusion of Innovation, Forecasting, Genomics trends, Humans, Medical Informatics trends, United States, American Heart Association, Biomedical Research trends, Cardiology trends
- Published
- 2017
- Full Text
- View/download PDF
28. Decreased KCNE2 Expression Participates in the Development of Cardiac Hypertrophy by Regulation of Calcineurin-NFAT (Nuclear Factor of Activated T Cells) and Mitogen-Activated Protein Kinase Pathways.
- Author
-
Liu W, Deng J, Ding W, Wang G, Shen Y, Zheng J, Zhang X, Luo Y, Lv C, Wang Y, Chen L, Yan D, Boudreau RL, Song LS, and Liu J
- Subjects
- Animals, Animals, Newborn, Apoptosis, Calcineurin metabolism, Cardiomegaly metabolism, Disease Models, Animal, Humans, Immunohistochemistry, Myocardium metabolism, Myocardium pathology, NFATC Transcription Factors metabolism, Polymerase Chain Reaction, Potassium Channels, Voltage-Gated biosynthesis, Rats, Rats, Sprague-Dawley, Calcineurin genetics, Cardiomegaly genetics, Gene Expression Regulation, Mitogen-Activated Protein Kinases metabolism, NFATC Transcription Factors genetics, Potassium Channels, Voltage-Gated genetics, RNA genetics
- Abstract
Background: KCNE2 is a promiscuous auxiliary subunit of voltage-gated cation channels. A recent work demonstrated that KCNE2 regulates L-type Ca
2+ channels. Given the important roles of altered Ca2+ signaling in structural and functional remodeling in diseased hearts, this study investigated whether KCNE2 participates in the development of pathological hypertrophy., Methods and Results: We found that cardiac KCNE2 expression was significantly decreased in phenylephrine-induced cardiomyocyte hypertrophy in neonatal rat ventricular myocytes and in transverse aortic constriction-induced cardiac hypertrophy in mice, as well as in dilated cardiomyopathy in human. Knockdown of KCNE2 in neonatal rat ventricular myocytes reproduced hypertrophy by increasing the expression of ANP (atrial natriuretic peptide) and β-MHC (β-myosin heavy chain), and cell surface area, whereas overexpression of KCNE2 attenuated phenylephrine-induced cardiomyocyte hypertrophy. Knockdown of KCNE2 increased intracellular Ca2+ transient, calcineurin activity, and nuclear NFAT (nuclear factor of activated T cells) protein levels, and pretreatment with inhibitor of L-type Ca2+ channel (nifedipine) or calcineurin (FK506) attenuated the activation of calcineurin-NFAT pathway and cardiomyocyte hypertrophy. Meanwhile, the phosphorylation levels of p38, extracellular signal-regulated kinase 1/2, and c-Jun N-terminal kinase were increased, and inhibiting the 3 cascades of mitogen-activated protein kinase reduced cardiomyocyte hypertrophy induced by KCNE2 knockdown. Overexpression of KCNE2 in heart by ultrasound-microbubble-mediated gene transfer suppressed the development of hypertrophy and activation of calcineurin-NFAT and mitogen-activated protein kinase pathways in transverse aortic constriction mice., Conclusions: This study demonstrates that cardiac KCNE2 expression is decreased and contributes to the development of hypertrophy via activation of calcineurin-NFAT and mitogen-activated protein kinase pathways. Targeting KCNE2 is a potential therapeutic strategy for the treatment of hypertrophy., (© 2017 American Heart Association, Inc.)- Published
- 2017
- Full Text
- View/download PDF
29. Sirtuin 1 regulates cardiac electrical activity by deacetylating the cardiac sodium channel.
- Author
-
Vikram A, Lewarchik CM, Yoon JY, Naqvi A, Kumar S, Morgan GM, Jacobs JS, Li Q, Kim YR, Kassan M, Liu J, Gabani M, Kumar A, Mehdi H, Zhu X, Guan X, Kutschke W, Zhang X, Boudreau RL, Dai S, Matasic DS, Jung SB, Margulies KB, Kumar V, Bachschmid MM, London B, and Irani K
- Subjects
- Acetylation, Animals, Echocardiography, Electrocardiography, HEK293 Cells, Heart diagnostic imaging, Heart physiopathology, Humans, Immunoblotting, Immunoprecipitation, Mass Spectrometry, Mice, Mice, Knockout, Myocytes, Cardiac, Patch-Clamp Techniques, Rats, Sirtuin 1 metabolism, Action Potentials, Arrhythmias, Cardiac genetics, Cardiomyopathies metabolism, Membrane Potentials, Myocardium metabolism, NAV1.5 Voltage-Gated Sodium Channel metabolism, Sirtuin 1 genetics
- Abstract
The voltage-gated cardiac Na
+ channel (Nav 1.5), encoded by the SCN5A gene, conducts the inward depolarizing cardiac Na+ current (INa ) and is vital for normal cardiac electrical activity. Inherited loss-of-function mutations in SCN5A lead to defects in the generation and conduction of the cardiac electrical impulse and are associated with various arrhythmia phenotypes. Here we show that sirtuin 1 deacetylase (Sirt1) deacetylates Nav 1.5 at lysine 1479 (K1479) and stimulates INa via lysine-deacetylation-mediated trafficking of Nav 1.5 to the plasma membrane. Cardiac Sirt1 deficiency in mice induces hyperacetylation of K1479 in Nav 1.5, decreases expression of Nav 1.5 on the cardiomyocyte membrane, reduces INa and leads to cardiac conduction abnormalities and premature death owing to arrhythmia. The arrhythmic phenotype of cardiac-Sirt1-deficient mice recapitulated human cardiac arrhythmias resulting from loss of function of Nav 1.5. Increased Sirt1 activity or expression results in decreased lysine acetylation of Nav 1.5, which promotes the trafficking of Nav 1.5 to the plasma membrane and stimulation of INa . As compared to wild-type Nav 1.5, Nav 1.5 with K1479 mutated to a nonacetylatable residue increases peak INa and is not regulated by Sirt1, whereas Nav 1.5 with K1479 mutated to mimic acetylation decreases INa . Nav 1.5 is hyperacetylated on K1479 in the hearts of patients with cardiomyopathy and clinical conduction disease. Thus, Sirt1, by deacetylating Nav 1.5, plays an essential part in the regulation of INa and cardiac electrical activity.- Published
- 2017
- Full Text
- View/download PDF
30. RNA Interference of Human α-Synuclein in Mouse.
- Author
-
Kim YC, Miller A, Lins LC, Han SW, Keiser MS, Boudreau RL, Davidson BL, and Narayanan NS
- Abstract
α-Synuclein is postulated to play a key role in the pathogenesis of Parkinson's disease (PD). Aggregates of α-synuclein contribute to neurodegeneration and cell death in humans and in mouse models of PD. Here, we use virally mediated RNA interference to knockdown human α-synuclein in mice. We used an siRNA design algorithm to identify eight siRNA sequences with minimal off-targeting potential. One RNA-interference sequence (miSyn4) showed maximal protein knockdown potential in vitro . We then designed AAV vectors expressing miSyn4 and injected them into the mouse substantia nigra. miSyn4 was robustly expressed and did not detectably change dopamine neurons, glial proliferation, or mouse behavior. We then injected AAV2-miSyn4 into Thy1-hSNCA mice over expressing α-synuclein and found decreased human α-synuclein (hSNCA) in both midbrain and cortex. In separate mice, co-injection of AAV2-hSNCA and AAV2-miSyn4 demonstrated decreased hSNCA expression and rescue of hSNCA-mediated behavioral deficits. These data suggest that virally mediated RNA interference can knockdown hSNCA in vivo , which could be helpful for future therapies targeting human α-synuclein.
- Published
- 2017
- Full Text
- View/download PDF
31. Elucidation of transcriptome-wide microRNA binding sites in human cardiac tissues by Ago2 HITS-CLIP.
- Author
-
Spengler RM, Zhang X, Cheng C, McLendon JM, Skeie JM, Johnson FL, Davidson BL, and Boudreau RL
- Subjects
- 3' Untranslated Regions genetics, Binding Sites, Calcium metabolism, Cardiomyopathies genetics, High-Throughput Nucleotide Sequencing, Humans, Myocardium cytology, Open Reading Frames genetics, Polymorphism, Single Nucleotide genetics, Substrate Specificity, Argonaute Proteins genetics, Argonaute Proteins metabolism, Cross-Linking Reagents, Immunoprecipitation, MicroRNAs metabolism, Myocardium metabolism, Transcriptome genetics
- Abstract
MicroRNAs (miRs) have emerged as key biological effectors in human health and disease. These small noncoding RNAs are incorporated into Argonaute (Ago) proteins, where they direct post-transcriptional gene silencing via base-pairing with target transcripts. Although miRs have become intriguing biological entities and attractive therapeutic targets, the translational impacts of miR research remain limited by a paucity of empirical miR targeting data, particularly in human primary tissues. Here, to improve our understanding of the diverse roles miRs play in cardiovascular function and disease, we applied high-throughput methods to globally profile miR:target interactions in human heart tissues. We deciphered Ago2:RNA interactions using crosslinking immunoprecipitation coupled with high-throughput sequencing (HITS-CLIP) to generate the first transcriptome-wide map of miR targeting events in human myocardium, detecting 4000 cardiac Ago2 binding sites across >2200 target transcripts. Our initial exploration of this interactome revealed an abundance of miR target sites in gene coding regions, including several sites pointing to new miR-29 functions in regulating cardiomyocyte calcium, growth and metabolism. Also, we uncovered several clinically-relevant interactions involving common genetic variants that alter miR targeting events in cardiomyopathy-associated genes. Overall, these data provide a critical resource for bolstering translational miR research in heart, and likely beyond., (© The Author(s) 2016. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2016
- Full Text
- View/download PDF
32. β-Secretase 1's Targeting Reduces Hyperphosphorilated Tau, Implying Autophagy Actors in 3xTg-AD Mice.
- Author
-
Piedrahita D, Castro-Alvarez JF, Boudreau RL, Villegas-Lanau A, Kosik KS, Gallego-Gomez JC, and Cardona-Gómez GP
- Abstract
β-site APP cleaving enzyme 1 (BACE1) initiates APP cleavage, which has been reported to be an inducer of tau pathology by altering proteasome functions in Alzheimer's disease (AD). However, the exact relationship between BACE1 and PHF (Paired Helical Filaments) formation is not clear. In this study, we confirm that BACE1 and Hsc70 are upregulated in the brains of AD patients, and we demonstrate that both proteins show enhanced expression in lipid rafts from AD-affected triple transgenic mouse brains. BACE1 targeting increased Hsc70 levels in the membrane and cytoplasm fractions and downregulated Hsp90 and CHIP in the nucleus in the hippocampi of 3xTg-AD mice. However, these observations occurred in a proteasome-independent manner in vitro. The BACE1miR-induced reduction of soluble hyperphosphorylated tau was associated with a decrease in MAPK activity. However, the BACE1 RNAi-mediated reduction of hyperphosphorylated tau was only blocked by 3-MA (3-methyladenine) in vitro, and it resulted in the increase of Hsc70 and LAMP2 in lipid rafts from hippocampi of 3xTg-AD mice, and upregulation of survival and homeostasis signaling. In summary, our findings suggest that BACE1 silencing neuroprotects reducing soluble hyperphosphorylated tau, modulating certain autophagy-related proteins in aged 3xTg-AD mice.
- Published
- 2016
- Full Text
- View/download PDF
33. Sonic Hedgehog Controls the Phenotypic Fate and Therapeutic Efficacy of Grafted Neural Precursor Cells in a Model of Nigrostriatal Neurodegeneration.
- Author
-
Madhavan L, Daley BF, Davidson BL, Boudreau RL, Lipton JW, Cole-Strauss A, Steece-Collier K, and Collier TJ
- Subjects
- Alkaline Phosphatase genetics, Alkaline Phosphatase metabolism, Animals, Animals, Newborn, Disease Models, Animal, Dopaminergic Neurons metabolism, Dopaminergic Neurons pathology, Doublecortin Protein, GPI-Linked Proteins genetics, GPI-Linked Proteins metabolism, Gene Expression Regulation, Developmental, Glial Cell Line-Derived Neurotrophic Factor antagonists & inhibitors, Glial Cell Line-Derived Neurotrophic Factor metabolism, Hedgehog Proteins antagonists & inhibitors, Hedgehog Proteins metabolism, Humans, Isoenzymes genetics, Isoenzymes metabolism, Mesencephalon growth & development, Mesencephalon metabolism, Mesencephalon pathology, Neostriatum growth & development, Neostriatum pathology, Neural Stem Cells cytology, Neural Stem Cells metabolism, Neurodegenerative Diseases chemically induced, Neurodegenerative Diseases metabolism, Neurodegenerative Diseases therapy, Neurogenesis genetics, Oxidopamine, Phenotype, Primary Cell Culture, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Rats, Rats, Inbred F344, Rats, Transgenic, Signal Transduction, Stereotaxic Techniques, Substantia Nigra growth & development, Substantia Nigra metabolism, Substantia Nigra pathology, Transgenes, Glial Cell Line-Derived Neurotrophic Factor genetics, Graft Survival genetics, Hedgehog Proteins genetics, Neostriatum metabolism, Neural Stem Cells transplantation, Neurodegenerative Diseases genetics
- Abstract
The expression of soluble growth and survival promoting factors by neural precursor cells (NPCs) is suggested to be a prominent mechanism underlying the protective and regenerative effects of these cells after transplantation. Nevertheless, how and to what extent specific NPC-expressed factors contribute to therapeutic effects is not well understood. Using RNA silencing, the current study investigated the roles of two donor NPC molecules, namely glial cell-line derived neurotrophic factor (GDNF) and sonic hedgehog (SHH), in the protection of substantia nigra dopamine neurons in rats treated with 6-hydroxydopamine (6-OHDA). Analyses indicate that as opposed to the knock-down of GDNF, SHH inhibition caused a profound decline in nigrostriatal neuroprotection. Further, SHH silencing also curbed endogenous neurogenesis and the migration of host brdU+/dcx+ neural precursors into the striatum, which was present in the animals receiving control or GDNF silenced NPCs. A change in graft phenotype, mainly reflected by a reduced proportion of undifferentiated nestin+ cells, as well as a significantly greater host microglial activity, suggested an important role for these processes in the attenuation of neuroprotection and neurogenesis upon SHH silencing. Overall these studies reveal core mechanisms fundamental to grafted NPC-based therapeutic effects, and delineate the particular contributions of two graft-expressed molecules, SHH and GDNF, in mediating midbrain dopamine neuron protection, and host plasticity after NPC transplantation.
- Published
- 2015
- Full Text
- View/download PDF
34. Inhibition of MCU forces extramitochondrial adaptations governing physiological and pathological stress responses in heart.
- Author
-
Rasmussen TP, Wu Y, Joiner ML, Koval OM, Wilson NR, Luczak ED, Wang Q, Chen B, Gao Z, Zhu Z, Wagner BA, Soto J, McCormick ML, Kutschke W, Weiss RM, Yu L, Boudreau RL, Abel ED, Zhan F, Spitz DR, Buettner GR, Song LS, Zingman LV, and Anderson ME
- Subjects
- Animals, Blood Pressure, Calcium metabolism, Cardiac Pacing, Artificial, Cellular Reprogramming, Cytosol drug effects, Cytosol metabolism, Diastole, Electrocardiography, Genes, Dominant, Glucose metabolism, Heart Ventricles pathology, Heart Ventricles physiopathology, Mice, Mitochondria, Heart drug effects, Myocardial Reperfusion, Myocardium metabolism, Myocardium pathology, Oxygen Consumption, Prostaglandin-Endoperoxide Synthases metabolism, Sarcoplasmic Reticulum metabolism, Transcription, Genetic, Adaptation, Physiological, Calcium Channels metabolism, Heart physiopathology, Mitochondria, Heart metabolism, Stress, Physiological
- Abstract
Myocardial mitochondrial Ca(2+) entry enables physiological stress responses but in excess promotes injury and death. However, tissue-specific in vivo systems for testing the role of mitochondrial Ca(2+) are lacking. We developed a mouse model with myocardial delimited transgenic expression of a dominant negative (DN) form of the mitochondrial Ca(2+) uniporter (MCU). DN-MCU mice lack MCU-mediated mitochondrial Ca(2+) entry in myocardium, but, surprisingly, isolated perfused hearts exhibited higher O2 consumption rates (OCR) and impaired pacing induced mechanical performance compared with wild-type (WT) littermate controls. In contrast, OCR in DN-MCU-permeabilized myocardial fibers or isolated mitochondria in low Ca(2+) were not increased compared with WT, suggesting that DN-MCU expression increased OCR by enhanced energetic demands related to extramitochondrial Ca(2+) homeostasis. Consistent with this, we found that DN-MCU ventricular cardiomyocytes exhibited elevated cytoplasmic [Ca(2+)] that was partially reversed by ATP dialysis, suggesting that metabolic defects arising from loss of MCU function impaired physiological intracellular Ca(2+) homeostasis. Mitochondrial Ca(2+) overload is thought to dissipate the inner mitochondrial membrane potential (ΔΨm) and enhance formation of reactive oxygen species (ROS) as a consequence of ischemia-reperfusion injury. Our data show that DN-MCU hearts had preserved ΔΨm and reduced ROS during ischemia reperfusion but were not protected from myocardial death compared with WT. Taken together, our findings show that chronic myocardial MCU inhibition leads to previously unanticipated compensatory changes that affect cytoplasmic Ca(2+) homeostasis, reprogram transcription, increase OCR, reduce performance, and prevent anticipated therapeutic responses to ischemia-reperfusion injury.
- Published
- 2015
- Full Text
- View/download PDF
35. Artificial miRNAs Targeting Mutant Huntingtin Show Preferential Silencing In Vitro and In Vivo.
- Author
-
Monteys AM, Wilson MJ, Boudreau RL, Spengler RM, and Davidson BL
- Abstract
Huntington's disease (HD) is a dominantly inherited neurodegenerative disease caused by CAG repeat expansion in exon 1 of huntingtin (HTT). Studies in mouse models of HD with a regulated mutant transgene show that continuous mutant allele expression is required for behavioral and pathological signs; when mutant HTT expression declined, neuronal degeneration improved. To date, it is unknown whether neural cells in the adult human brain can tolerate reduction in both normal and mutant alleles. Thus, it may be important to develop allele-specific silencing approaches. Several siRNA sequences targeting the CAG expanded motif or prevalent single-nucleotide polymorphisms (SNPs) in linkage disequilibrium with the mutant allele have been designed and their selectivity demonstrated in vitro. However, it is unknown whether these allele-specific siRNAs will retain their specificity when expressed from artificial RNAi platforms. Here, we designed CAG- and SNP- targeting artificial miRNAs and demonstrate that some, but not all, retained their selectivity in vitro using an allele-specific reporter system and in vivo in a transgenic mouse model developed to express normal and mutant human HTT alleles.
- Published
- 2015
- Full Text
- View/download PDF
36. Nonallele specific silencing of ataxin-7 improves disease phenotypes in a mouse model of SCA7.
- Author
-
Ramachandran PS, Boudreau RL, Schaefer KA, La Spada AR, and Davidson BL
- Subjects
- Alleles, Animals, Ataxin-7, Disease Models, Animal, Gene Expression Regulation, Mice, Mice, Inbred C57BL, Motor Activity, Mutation, Purkinje Cells pathology, Spinocerebellar Ataxias genetics, Nerve Tissue Proteins antagonists & inhibitors, Nerve Tissue Proteins genetics, Purkinje Cells metabolism, RNA Interference, Spinocerebellar Ataxias physiopathology, Spinocerebellar Ataxias therapy
- Abstract
Spinocerebellar ataxia type 7 (SCA7) is a late-onset neurodegenerative disease characterized by ataxia and vision loss with no effective treatments in the clinic. The most striking feature is the degeneration of Purkinje neurons of the cerebellum caused by the presence of polyglutamine-expanded ataxin-7. Ataxin-7 is part of a transcriptional complex, and, in the setting of mutant ataxin-7, there is misregulation of target genes. Here, we designed RNAi sequences to reduce the expression of both wildtype and mutant ataxin-7 to test if reducing ataxin-7 in Purkinje cells is both tolerated and beneficial in an animal model of SCA7. We observed sustained reduction of both wildtype and mutant ataxin-7 as well as a significant improvement of ataxia phenotypes. Furthermore, we observed a reduction in cerebellar molecular layer thinning and nuclear inclusions, a hallmark of SCA7. In addition, we observed recovery of cerebellar transcripts whose expression is disrupted in the presence of mutant ataxin-7. These data demonstrate that reduction of both wildtype and mutant ataxin-7 by RNAi is well tolerated, and contrary to what may be expected from reducing a component of the Spt-Taf9-Gcn5 acetyltransferase complex, is efficacious in the SCA7 mouse.
- Published
- 2014
- Full Text
- View/download PDF
37. A microRNA processing defect in smokers' macrophages is linked to SUMOylation of the endonuclease DICER.
- Author
-
Gross TJ, Powers LS, Boudreau RL, Brink B, Reisetter A, Goel K, Gerke AK, Hassan IH, and Monick MM
- Subjects
- Acetylcysteine pharmacology, Blotting, Western, Down-Regulation, Free Radical Scavengers pharmacology, HeLa Cells, Humans, Oligonucleotide Array Sequence Analysis, RNA Interference, Reverse Transcriptase Polymerase Chain Reaction, Smoke, Sumoylation drug effects, Nicotiana chemistry, Transcriptome, Ubiquitin-Conjugating Enzymes genetics, Ubiquitin-Conjugating Enzymes metabolism, Ubiquitin-Conjugating Enzyme UBC9, DEAD-box RNA Helicases metabolism, Macrophages, Alveolar metabolism, MicroRNAs genetics, Ribonuclease III metabolism, Smoking
- Abstract
Despite the fact that alveolar macrophages play an important role in smoking-related disease, little is known about what regulates their pathophysiologic phenotype. Evaluating smoker macrophages, we found significant down-regulation of multiple microRNAs (miRNAs). This work investigates the hypothesis that cigarette smoke alters mature miRNA expression in lung macrophages by inhibiting processing of primary miRNA transcripts. Studies on smoker alveolar macrophages showed a defect in miRNA maturation. Studies on the miRNA biogenesis machinery led us to focus on the cytosolic RNA endonuclease, DICER. DICER cleaves the stem-loop structure from pre-miRNAs, allowing them to dissociate into their mature 20-22-nucleotide single-stranded form. DICER activity assays confirmed impaired DICER activity following cigarette smoke exposure. Further protein studies demonstrated a decreased expression of the native 217-kDa form of DICER and an accumulation of high molecular weight forms with cigarette smoke exposure. This molecular mass shift was shown to contain SUMO moieties and could be blocked by silencing RNA directed at the primary SUMOylating ligase, Ubc9. In determining the cigarette smoke components responsible for changes in DICER, we found that N-acetylcysteine, an antioxidant and anti-aldehyde, protected DICER protein and activity from cigarette smoke extract. This massive down-regulation of miRNAs (driven in part by alterations in DICER) may be an important regulator of the disease-promoting macrophage phenotype found in the lungs of smokers.
- Published
- 2014
- Full Text
- View/download PDF
38. RNA interference-based therapy for spinocerebellar ataxia type 7 retinal degeneration.
- Author
-
Ramachandran PS, Bhattarai S, Singh P, Boudreau RL, Thompson S, Laspada AR, Drack AV, and Davidson BL
- Subjects
- Animals, Ataxin-7, Humans, Mice, Mice, Inbred C57BL, Mice, Transgenic, Nerve Tissue Proteins genetics, Retinal Degeneration genetics, RNA Interference physiology, Retinal Degeneration therapy, Spinocerebellar Ataxias genetics, Spinocerebellar Ataxias therapy
- Abstract
Spinocerebellar ataxia type 7 (SCA7) is an autosomal dominant neurodegenerative disease characterized by loss of motor coordination and retinal degeneration with no current therapies in the clinic. The causative mutation is an expanded CAG repeat in the ataxin-7 gene whose mutant protein product causes cerebellar and brainstem degeneration and retinal cone-rod dystrophy. Here, we reduced the expression of both mutant and wildtype ataxin-7 in the SCA7 mouse retina by RNA interference and evaluated retinal function 23 weeks post injection. We observed a preservation of normal retinal function and no adverse toxicity with ≥50% reduction of mutant and wildtype ataxin-7 alleles. These studies address an important safety concern regarding non-allele specific silencing of ataxin-7 for SCA7 retinal therapy.
- Published
- 2014
- Full Text
- View/download PDF
39. Broad therapeutic benefit after RNAi expression vector delivery to deep cerebellar nuclei: implications for spinocerebellar ataxia type 1 therapy.
- Author
-
Keiser MS, Boudreau RL, and Davidson BL
- Subjects
- Animals, Ataxin-1, Ataxins, Dependovirus genetics, Disease Models, Animal, Female, Gene Knock-In Techniques, Humans, Male, Mice, Mice, Inbred C57BL, Spinocerebellar Ataxias genetics, Spinocerebellar Ataxias therapy, Cerebellar Cortex metabolism, Cerebellar Nuclei virology, Genetic Vectors administration & dosage, Nerve Tissue Proteins antagonists & inhibitors, Nerve Tissue Proteins genetics, Neurons metabolism, Nuclear Proteins antagonists & inhibitors, Nuclear Proteins genetics, RNA Interference, Spinocerebellar Ataxias pathology
- Abstract
Spinocerebellar ataxia type 1 (SCA1) is an autosomal dominant, late-onset neurodegenerative disease caused by a polyglutamine (polyQ) expansion in the ataxin-1 protein, which causes progressive neurodegeneration in cerebellar Purkinje cells and brainstem nuclei. Here, we tested if reducing mutant ataxin-1 expression would significantly improve phenotypes in a knock-in (KI) mouse model that recapitulates spatial and temporal aspects of SCA1. Adeno-associated viruses (AAVs), expressing inhibitory RNAs targeting ataxin-1, were injected into the deep cerebellar nuclei (DCN) of KI mice. This approach induced ataxin-1 suppression in the cerebellar cortex and in brainstem neurons. RNA interference (RNAi) of ataxin-1 preserved cerebellar lobule integrity and prevented disease-related transcriptional changes for over a year. Notably, RNAi therapy also preserved rotarod performance and neurohistology. These data suggest that delivery of AAVs encoding RNAi sequences against ataxin-1, to DCN alone, may be sufficient for SCA1 therapy.
- Published
- 2014
- Full Text
- View/download PDF
40. Transcriptome-wide discovery of microRNA binding sites in human brain.
- Author
-
Boudreau RL, Jiang P, Gilmore BL, Spengler RM, Tirabassi R, Nelson JA, Ross CA, Xing Y, and Davidson BL
- Subjects
- Adult, Aged, Animals, Argonaute Proteins genetics, Argonaute Proteins metabolism, Autoradiography, Base Sequence, Gene Regulatory Networks, Humans, Immunoprecipitation, Male, Mice, Middle Aged, Motor Cortex cytology, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Postmortem Changes, RNA, Messenger, Transcriptome physiology, Binding Sites genetics, Gyrus Cinguli metabolism, MicroRNAs genetics, MicroRNAs metabolism, Motor Cortex metabolism
- Abstract
The orchestration of brain function requires complex gene regulatory networks that are modulated, in part, by microRNAs (miRNAs). These noncoding RNAs associate with argonaute (Ago) proteins in order to direct posttranscriptional gene suppression via base pairing with target transcripts. In order to better understand how miRNAs contribute to human-specialized brain processes and neurological phenotypes, identifying their targets is of paramount importance. Here, we address the latter by profiling Ago2:RNA interactions using HITS-CLIP to generate a transcriptome-wide map of miRNA binding sites in human brain. We uncovered ∼ 7,000 stringent Ago2 binding sites that are highly enriched for conserved sequences corresponding to abundant brain miRNAs. This interactome points to functional miRNA:target pairs across >3,000 genes and represents a valuable resource for accelerating our understanding of miRNA functions in brain. We demonstrate the utility of this map for exploring clinically relevant miRNA binding sites that may facilitate the translation of genetic studies of complex neuropsychiatric diseases into therapeutics., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
41. Rhes suppression enhances disease phenotypes in Huntington's disease mice.
- Author
-
Lee JH, Sowada MJ, Boudreau RL, Aerts AM, Thedens DR, Nopoulos P, and Davidson BL
- Subjects
- Animals, Anxiety metabolism, Atrophy, Behavior, Animal, Disease Models, Animal, Genetic Therapy, Huntingtin Protein, Huntington Disease genetics, Magnetic Resonance Imaging, Phenotype, GTP-Binding Proteins antagonists & inhibitors, Huntington Disease metabolism, MicroRNAs, Mutant Proteins metabolism, Neostriatum pathology, Nerve Tissue Proteins metabolism, Nuclear Proteins metabolism
- Abstract
In Huntington's disease (HD) mutant HTT is ubiquitously expressed yet the striatum undergoes profound early degeneration. Cell culture studies suggest that a striatal-enriched protein, Rhes, may account for this vulnerability. We investigated the therapeutic potential of silencing Rhes in vivo using inhibitory RNAs (miRhes). While Rhes suppression was tolerated in wildtype mice, it failed to improve rotarod function in two distinct HD mouse models. Additionally, miRhes treated HD mice had increased anxiety-like behaviors and enhanced striatal atrophy as measured by longitudinal MRI when compared to control treated mice. These findings raise caution regarding the long-term implementation of inhibiting Rhes as a therapy for HD.
- Published
- 2014
- Full Text
- View/download PDF
42. Silencing mutant ATXN3 expression resolves molecular phenotypes in SCA3 transgenic mice.
- Author
-
Rodríguez-Lebrón E, Costa Mdo C, Luna-Cancalon K, Peron TM, Fischer S, Boudreau RL, Davidson BL, and Paulson HL
- Subjects
- 3' Untranslated Regions, Animals, Ataxin-3, Cerebellum pathology, Dependovirus drug effects, Dependovirus genetics, Disease Models, Animal, Gene Expression Regulation, Gene Silencing, Genetic Vectors drug effects, Genetic Vectors genetics, HEK293 Cells, Humans, Machado-Joseph Disease genetics, Mice, Mice, Transgenic, MicroRNAs pharmacology, Molecular Mimicry, Molecular Targeted Therapy, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Nuclear Proteins genetics, Nuclear Proteins metabolism, Repressor Proteins genetics, Repressor Proteins metabolism, Transduction, Genetic methods, Machado-Joseph Disease pathology, MicroRNAs adverse effects, Mutant Proteins drug effects, Nerve Tissue Proteins drug effects, Nuclear Proteins drug effects, Phenotype, Repressor Proteins drug effects
- Abstract
Spinocerebellar ataxia type 3 (SCA3) is a neurodegenerative disease caused by a polyglutamine expansion in the deubiquitinating enzyme, Ataxin-3. Currently, there are no effective treatments for this fatal disorder but studies support the hypothesis that reducing mutant Ataxin-3 protein levels might reverse or halt the progression of disease in SCA3. Here, we sought to modulate ATXN3 expression in vivo using RNA interference. We developed artificial microRNA mimics targeting the 3'-untranslated region (3'UTR) of human ATXN3 and then used recombinant adeno-associated virus to deliver them to the cerebellum of transgenic mice expressing the full human disease gene (SCA3/MJD84.2 mice). Anti-ATXN3 microRNA mimics effectively suppressed human ATXN3 expression in SCA3/MJD84.2 mice. Short-term treatment cleared the abnormal nuclear accumulation of mutant Ataxin-3 throughout the transduced SCA3/MJD84.2 cerebellum. Analysis also revealed changes in the steady-state levels of specific microRNAs in the cerebellum of SCA3/MJD84.2 mice, a previously uncharacterized molecular phenotype of SCA3 that appears to be dependent on mutant Ataxin-3 expression. Our findings support the preclinical development of molecular therapies aimed at halting the expression of ATXN3 as a viable approach to SCA3 and point to microRNA deregulation as a potential surrogate marker of SCA3 pathogenesis.
- Published
- 2013
- Full Text
- View/download PDF
43. RNAi or overexpression: alternative therapies for Spinocerebellar Ataxia Type 1.
- Author
-
Keiser MS, Geoghegan JC, Boudreau RL, Lennox KA, and Davidson BL
- Subjects
- Animals, Ataxin-1, Ataxins, Behavior, Animal physiology, Blotting, Western, Brain pathology, Dependovirus genetics, Gait physiology, Genetic Vectors, HEK293 Cells, Humans, Immunohistochemistry, Immunoprecipitation, In Situ Hybridization, Locomotion physiology, Mice, Mice, Transgenic, MicroRNAs biosynthesis, MicroRNAs genetics, Nerve Tissue Proteins genetics, Nuclear Proteins genetics, Plasmids, Postural Balance physiology, RNA, Small Interfering therapeutic use, Real-Time Polymerase Chain Reaction, Spinocerebellar Ataxias pathology, Spinocerebellar Ataxias psychology, Nerve Tissue Proteins biosynthesis, Nuclear Proteins biosynthesis, RNA Interference physiology, Spinocerebellar Ataxias therapy
- Abstract
Spinocerebellar Ataxia Type 1 (SCA1) is an autosomal dominant late onset neurodegenerative disease caused by an expanded polyglutamine tract in ataxin-1. Here, we compared the protective effects of overexpressing ataxin-1-like using recombinant AAVs, or reducing expression of mutant ataxin-1 using virally delivered RNA interference (RNAi), in a transgenic mouse model of SCA1. For the latter, we used an artificial microRNA (miR) design that optimizes potency, efficacy and safety to suppress ataxin-1 expression (miS1). Delivery of either ataxin-1-like or miS1 viral vectors to SCA1 mice cerebella resulted in widespread cerebellar Purkinje cell transduction and improved behavioral and histological phenotypes. Our data indicate the utility of either approach as a possible therapy for SCA1 patients., (Published by Elsevier Inc.)
- Published
- 2013
- Full Text
- View/download PDF
44. siSPOTR: a tool for designing highly specific and potent siRNAs for human and mouse.
- Author
-
Boudreau RL, Spengler RM, Hylock RH, Kusenda BJ, Davis HA, Eichmann DA, and Davidson BL
- Subjects
- Algorithms, Animals, Cell Line, Genome, Humans, Mice, Transcriptome, RNA Interference, RNA, Small Interfering chemistry, Software
- Abstract
RNA interference (RNAi) serves as a powerful and widely used gene silencing tool for basic biological research and is being developed as a therapeutic avenue to suppress disease-causing genes. However, the specificity and safety of RNAi strategies remains under scrutiny because small inhibitory RNAs (siRNAs) induce off-target silencing. Currently, the tools available for designing siRNAs are biased toward efficacy as opposed to specificity. Prior work from our laboratory and others' supports the potential to design highly specific siRNAs by limiting the promiscuity of their seed sequences (positions 2-8 of the small RNA), the primary determinant of off-targeting. Here, a bioinformatic approach to predict off-targeting potentials was established using publically available siRNA data from more than 50 microarray experiments. With this, we developed a specificity-focused siRNA design algorithm and accompanying online tool which, upon validation, identifies candidate sequences with minimal off-targeting potentials and potent silencing capacities. This tool offers researchers unique functionality and output compared with currently available siRNA design programs. Furthermore, this approach can greatly improve genome-wide RNAi libraries and, most notably, provides the only broadly applicable means to limit off-targeting from RNAi expression vectors.
- Published
- 2013
- Full Text
- View/download PDF
45. Generation of hairpin-based RNAi vectors for biological and therapeutic application.
- Author
-
Boudreau RL and Davidson BL
- Subjects
- Animals, Base Sequence, Cloning, Molecular methods, Humans, MicroRNAs genetics, MicroRNAs isolation & purification, Molecular Sequence Data, Polymerase Chain Reaction methods, Viruses genetics, Genetic Therapy methods, Genetic Vectors, RNA Interference, RNA, Small Interfering genetics
- Abstract
RNA interference (RNAi) is a natural process of gene silencing mediated by small RNAs. Shortly after the discovery of the RNAi mechanism, scientists devised various methods of delivering small interfering RNAs (siRNAs) capable of co-opting the endogenous RNAi machinery and suppressing target gene expression based on sequence complementarity. RNAi has since become a powerful tool to study gene function and is being investigated as a potential therapeutic approach to treat a vast array of human diseases (e.g., cancer, viral infections, and dominant genetic disorders). Among the available RNAi vectors are hairpin-based expression platforms (short-hairpin RNAs and artificial microRNAs) designed to mimic endogenously expressed inhibitory RNAs. These RNAi vectors are capable of achieving long-term potent gene silencing in vitro and in vivo. Here, we describe methods to design and generate these hairpin-based vectors and briefly review considerations for downstream applications., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
46. Preclinical safety of RNAi-mediated HTT suppression in the rhesus macaque as a potential therapy for Huntington's disease.
- Author
-
McBride JL, Pitzer MR, Boudreau RL, Dufour B, Hobbs T, Ojeda SR, and Davidson BL
- Subjects
- Animals, Behavior, Animal, Blotting, Western, Dependovirus genetics, Drug Evaluation, Preclinical, Gliosis metabolism, Gliosis pathology, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Humans, Huntingtin Protein, Immunity, Active, Immunoenzyme Techniques, Inflammation metabolism, Inflammation pathology, Macaca mulatta, Magnetic Resonance Imaging, Male, MicroRNAs administration & dosage, MicroRNAs genetics, Motor Activity, Nerve Tissue Proteins metabolism, Neurons metabolism, Neurons pathology, Nuclear Proteins metabolism, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Huntington Disease genetics, Huntington Disease therapy, Nerve Tissue Proteins chemistry, Nerve Tissue Proteins genetics, Nuclear Proteins chemistry, Nuclear Proteins genetics, RNA Interference, RNA, Small Interfering genetics
- Abstract
To date, a therapy for Huntington's disease (HD), a genetic, neurodegenerative disorder, remains elusive. HD is characterized by cell loss in the basal ganglia, with particular damage to the putamen, an area of the brain responsible for initiating and refining motor movements. Consequently, patients exhibit a hyperkinetic movement disorder. RNA interference (RNAi) offers therapeutic potential for this disorder by reducing the expression of HTT, the disease-causing gene. We have previously demonstrated that partial suppression of both wild-type and mutant HTT in the striatum prevents behavioral and neuropathological abnormalities in rodent models of HD. However, given the role of HTT in various cellular processes, it remains unknown whether a partial suppression of both alleles will be safe in mammals whose neurophysiology, basal ganglia anatomy, and behavioral repertoire more closely resembles that of a human. Here, we investigate whether a partial reduction of HTT in the normal non-human primate putamen is safe. We demonstrate that a 45% reduction of rhesus HTT expression in the mid- and caudal putamen does not induce motor deficits, neuronal degeneration, astrogliosis, or an immune response. Together, these data suggest that partial suppression of wild-type HTT expression is well tolerated in the primate putamen and further supports RNAi as a therapy for HD.
- Published
- 2011
- Full Text
- View/download PDF
47. Rational design of therapeutic siRNAs: minimizing off-targeting potential to improve the safety of RNAi therapy for Huntington's disease.
- Author
-
Boudreau RL, Spengler RM, and Davidson BL
- Subjects
- Animals, Blotting, Western, Brain metabolism, Brain pathology, Corpus Striatum metabolism, Corpus Striatum pathology, Dependovirus genetics, Drug Design, Gene Targeting, Humans, Immunoenzyme Techniques, Mice, MicroRNAs administration & dosage, MicroRNAs genetics, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Serotonin Plasma Membrane Transport Proteins metabolism, Gene Silencing, Huntington Disease genetics, Huntington Disease therapy, RNA Interference, RNA, Small Interfering genetics, Serotonin Plasma Membrane Transport Proteins chemistry, Serotonin Plasma Membrane Transport Proteins genetics
- Abstract
RNA interference (RNAi) provides an approach for the treatment of many human diseases. However, the safety of RNAi-based therapies can be hampered by the ability of small inhibitory RNAs (siRNAs) to bind to unintended mRNAs and reduce their expression, an effect known as off-target gene silencing. Off-targeting primarily occurs when the seed region (nucleotides 2-8 of the small RNA) pairs with sequences in 3'-UTRs of unintended mRNAs and directs translational repression and destabilization of those transcripts. To date, most therapeutic RNAi sequences are selected primarily for gene silencing efficacy, and later evaluated for safety. Here, in designing siRNAs to treat Huntington's disease (HD), a dominant neurodegenerative disorder, we prioritized selection of sequences with minimal off-targeting potentials (i.e., those with a scarcity of seed complements within all known human 3'-UTRs). We identified new promising therapeutic candidate sequences which show potent silencing in cell culture and mouse brain. Furthermore, we present microarray data demonstrating that off-targeting is significantly minimized by using siRNAs that contain "safe" seeds, an important strategy to consider during preclinical development of RNAi-based therapeutics.
- Published
- 2011
- Full Text
- View/download PDF
48. RNAi medicine for the brain: progresses and challenges.
- Author
-
Boudreau RL, Rodríguez-Lebrón E, and Davidson BL
- Subjects
- Brain pathology, Central Nervous System, Gene Silencing, Humans, Models, Biological, Neurodegenerative Diseases genetics, Peptides genetics, Genetic Therapy methods, Neurodegenerative Diseases therapy, RNA Interference
- Abstract
RNAi interference (RNAi) is a powerful gene silencing technology that has immense potential for treating a vast array of human ailments, for which suppressing disease-associated genes may provide clinical benefit. Here, we review the development of RNAi as a therapeutic modality for neurodegenerative diseases affecting the central nervous system (CNS). We overview promising preclinical data for the application of RNAi in the CNS and discuss key challenges (e.g. delivery and specificity) that remain as these approaches transition to the clinic.
- Published
- 2011
- Full Text
- View/download PDF
49. Control of hepatic nuclear superoxide production by glucose 6-phosphate dehydrogenase and NADPH oxidase-4.
- Author
-
Spencer NY, Yan Z, Boudreau RL, Zhang Y, Luo M, Li Q, Tian X, Shah AM, Davisson RL, Davidson B, Banfi B, and Engelhardt JF
- Subjects
- Animals, Cell Nucleus genetics, Chickens, Electron Spin Resonance Spectroscopy, Enzyme Activators metabolism, Glucosephosphate Dehydrogenase genetics, Liver cytology, Mice, Mice, Knockout, NADP metabolism, NADPH Oxidase 4, NADPH Oxidases genetics, Nuclear Proteins genetics, Rabbits, Cell Nucleus metabolism, Glucosephosphate Dehydrogenase metabolism, Liver enzymology, NADPH Oxidases metabolism, Nuclear Proteins metabolism, Superoxides metabolism
- Abstract
Redox-regulated signal transduction is coordinated by spatially controlled production of reactive oxygen species within subcellular compartments. The nucleus has long been known to produce superoxide (O(2)(·-)); however, the mechanisms that control this function remain largely unknown. We have characterized molecular features of a nuclear superoxide-producing system in the mouse liver. Using electron paramagnetic resonance, we investigated whether several NADPH oxidases (NOX1, 2, and 4) and known activators of NOX (Rac1, Rac2, p22(phox), and p47(phox)) contribute to nuclear O(2)(·-) production in isolated hepatic nuclei. Our findings demonstrate that NOX4 most significantly contributes to hepatic nuclear O(2)(·-) production that utilizes NADPH as an electron donor. Although NOX4 protein immunolocalized to both nuclear membranes and intranuclear inclusions, fluorescent detection of NADPH-dependent nuclear O(2)(·-) predominantly localized to the perinuclear space. Interestingly, NADP(+) and G6P also induced nuclear O(2)(·-) production, suggesting that intranuclear glucose-6-phosphate dehydrogenase (G6PD) can control NOX4 activity through nuclear NADPH production. Using G6PD mutant mice and G6PD shRNA, we confirmed that reductions in nuclear G6PD enzyme decrease the ability of hepatic nuclei to generate O(2)(·-) in response to NADP(+) and G6P. NOX4 and G6PD protein were also observed in overlapping microdomains within the nucleus. These findings provide new insights on the metabolic pathways for substrate regulation of nuclear O(2)(·-) production by NOX4.
- Published
- 2011
- Full Text
- View/download PDF
50. Silencing of CDK5 as potential therapy for Alzheimer's disease.
- Author
-
López-Tobón A, Castro-Álvarez JF, Piedrahita D, Boudreau RL, Gallego-Gómez JC, and Cardona-Gómez GP
- Subjects
- Animals, Humans, Models, Biological, Neurodegenerative Diseases enzymology, Phosphorylation, Alzheimer Disease enzymology, Alzheimer Disease therapy, Cyclin-Dependent Kinase 5 metabolism, RNA Interference physiology
- Abstract
Neurodegeneration is one of the greatest public health challenges for the 21st century. Among neurodegenerative diseases, Alzheimer's disease (AD) is the most prevalent and best characterized. Nevertheless, despite the large investment in AD research, currently there is no effective therapeutic option. In the present review, we highlight a novel alternative, which takes advantage of the biotechnological outbreak deployed by the discovery of the RNA interference-based gene silencing mechanism, and its application as a tool for neurodegeneration treatment. Here, we highlight cyclin-dependent kinase 5 (CDK5) as a key candidate target for therapeutic gene silencing. Unlike other members of the cyclin-dependent kinase family, CDK5 does not seem to play a crucial role in cell cycle regulation. By contrast, CDK5 participates in multiple functions during nervous system development and has been established as a key mediator of Tau hyperphosphorylation and neurofibrillary pathology, thus serving as an optimal candidate for targeted therapy in the adult nervous system. We propose that the use of RNA interference for CDK5 silencing presents an attractive and specific therapeutic alternative for AD and perhaps against other tauopathies.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.