301
|
Tu WY, Simpson JE, Highley JR, Heath PR. Spinal muscular atrophy: Factors that modulate motor neurone vulnerability. Neurobiol Dis 2017; 102:11-20. [PMID: 28161391 DOI: 10.1016/j.nbd.2017.01.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/10/2017] [Accepted: 01/31/2017] [Indexed: 01/07/2023] Open
Abstract
Spinal muscular atrophy (SMA), a leading genetic cause of infant death, is a neurodegenerative disease characterised by the selective loss of particular groups of motor neurones in the anterior horn of the spinal cord with concomitant muscle weakness. To date, no effective treatment is available, however, there are ongoing clinical trials are in place which promise much for the future. However, there remains an ongoing problem in trying to link a single gene loss to motor neurone degeneration. Fortunately, given successful disease models that have been established and intensive studies on SMN functions in the past ten years, we are fast approaching the stage of identifying the underlying mechanisms of SMA pathogenesis Here we discuss potential disease modifying factors on motor neurone vulnerability, in the belief that these factors give insight into the pathological mechanisms of SMA and therefore possible therapeutic targets.
Collapse
Affiliation(s)
- Wen-Yo Tu
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Julie E Simpson
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - J Robin Highley
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Paul R Heath
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK.
| |
Collapse
|
302
|
Ahfeldt T, Litterman NK, Rubin LL. Studying human disease using human neurons. Brain Res 2017; 1656:40-48. [PMID: 27060768 PMCID: PMC5053850 DOI: 10.1016/j.brainres.2016.03.051] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 03/08/2016] [Accepted: 03/31/2016] [Indexed: 01/25/2023]
Abstract
Utilizing patient derived cells has enormous promise for discovering new drugs for diseases of the nervous system, a goal that has been historically quite challenging. In this review, we will outline the potential of human stem cell derived neuron models for assessing therapeutics and high-throughput screening and compare to more traditional drug discovery strategies. We summarize recent successes of the approach and discuss special considerations for developing human stem cell based assays. New technologies, such as genome editing, offer improvements to help overcome the challenges that remain. Finally, human neurons derived from patient cells have advantages for translational research beyond drug screening as they can also be used to identify individual efficacy and safety prior to clinical testing and for dissecting disease mechanisms. This article is part of a Special Issue entitled SI: Exploiting human neurons.
Collapse
Affiliation(s)
- Tim Ahfeldt
- Department of Stem Cells and Regenerative Biology, Harvard University, Cambridge MA , USA, , Fax: 617-495-3961
| | - Nadia K. Litterman
- Department of Stem Cells and Regenerative Biology, Harvard University, Cambridge MA , USA, , Fax: 617-495-3961
| | - Lee L. Rubin
- Department of Stem Cells and Regenerative Biology, Harvard University, Cambridge MA , USA, , Fax: 617-495-3961
| |
Collapse
|
303
|
Shi Y, Inoue H, Wu JC, Yamanaka S. Induced pluripotent stem cell technology: a decade of progress. Nat Rev Drug Discov 2017; 16:115-130. [PMID: 27980341 PMCID: PMC6416143 DOI: 10.1038/nrd.2016.245] [Citation(s) in RCA: 1005] [Impact Index Per Article: 125.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since the advent of induced pluripotent stem cell (iPSC) technology a decade ago, enormous progress has been made in stem cell biology and regenerative medicine. Human iPSCs have been widely used for disease modelling, drug discovery and cell therapy development. Novel pathological mechanisms have been elucidated, new drugs originating from iPSC screens are in the pipeline and the first clinical trial using human iPSC-derived products has been initiated. In particular, the combination of human iPSC technology with recent developments in gene editing and 3D organoids makes iPSC-based platforms even more powerful in each area of their application, including precision medicine. In this Review, we discuss the progress in applications of iPSC technology that are particularly relevant to drug discovery and regenerative medicine, and consider the remaining challenges and the emerging opportunities in the field.
Collapse
Affiliation(s)
- Yanhong Shi
- Division of Stem Cell Biology Research, Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, California 91010, USA
| | - Haruhisa Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Joseph C Wu
- Stanford Cardiovascular Institute, 265 Campus Drive, Room G1120B, Stanford, California 94305-5454, USA
| | - Shinya Yamanaka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
- Gladstone Institute of Cardiovascular Disease, San Francisco, California 94158, USA
| |
Collapse
|
304
|
Securinine enhances SMN2 exon 7 inclusion in spinal muscular atrophy cells. Biomed Pharmacother 2017; 88:708-714. [PMID: 28152480 DOI: 10.1016/j.biopha.2017.01.104] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/14/2017] [Accepted: 01/17/2017] [Indexed: 11/23/2022] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive disease characterized by the degeneration of motor neurons in the spinal cord, leading to muscular atrophy. SMA is caused by deletions or mutations in the survival motor neuron gene (SMN1) on chromosome 5q13. A second copy of the SMN gene (SMN2) also exists on chromosome 5, and both genes can produce functional protein. However, due to alternative splicing of the exon 7, the majority of SMN protein produced by SMN2 is truncated and unable to compensate for the loss of SMN1. Increasing full-length SMN protein production by promoting the exon 7 inclusion in SMN2 mRNA or increasing SMN2 gene transcription could be a therapeutic approach for SMA. In this study, we screened for the compounds that enhance SMN2 exon 7 inclusion by using SMN2 minigene-luciferase reporter system. We found that securinine can increase luciferase activity, indicating that securinine promoted SMN2 exon 7 inclusion. In addition, securinine increased full-length SMN2 mRNA and SMN protein expression in SMA patient-derived lymphoid cell lines. To investigate the mechanism of securinine effect on SMN2 splicing, we compared the protein levels of relevant splicing factors between securinine-treated and untreated cells. We found that securinine downregulated hnRNP A1 and Sam68 and upregulated Tra2-β1 expression. However, securinine, unlike HDAC inhibitors, did not enhance tra2-β1 gene transcription, indicating a post-transcriptional mechanism for Tra2-β1 upregulation. Furthermore, we treated SMA-like mice with securinine by i.p. injection and found that securinine treatment increased SMN2 exon 7 inclusion and SMN protein expression in the brain and spinal cord. According to our results, securinine might have the potential to become a therapeutic drug for SMA disease.
Collapse
|
305
|
LOX-1 and Its Splice Variants: A New Challenge for Atherosclerosis and Cancer-Targeted Therapies. Int J Mol Sci 2017; 18:ijms18020290. [PMID: 28146073 PMCID: PMC5343826 DOI: 10.3390/ijms18020290] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/15/2017] [Accepted: 01/23/2017] [Indexed: 12/13/2022] Open
Abstract
Alternative splicing (AS) is a process in which precursor messenger RNA (pre-mRNA) splicing sites are differentially selected to diversify the protein isoform population. Changes in AS patterns have an essential role in normal development, differentiation and response to physiological stimuli. It is documented that AS can generate both “risk” and “protective” splice variants that can contribute to the pathogenesis of several diseases including atherosclerosis. The main endothelial receptor for oxidized low-density lipoprotein (ox-LDLs) is LOX-1 receptor protein encoded by the OLR1 gene. When OLR1 undergoes AS events, it generates three variants: OLR1, OLR1D4 and LOXIN. The latter lacks exon 5 and two-thirds of the functional domain. Literature data demonstrate a protective role of LOXIN in pathologies correlated with LOX-1 overexpression such as atherosclerosis and tumors. In this review, we summarize recent developments in understanding of OLR1 AS while also highlighting data warranting further investigation of this process as a novel therapeutic target.
Collapse
|
306
|
Sigala I, Ganidis G, Thysiadis S, Zografos AL, Giannakouros T, Sarli V, Nikolakaki E. Lynamicin D an antimicrobial natural product affects splicing by inducing the expression of SR protein kinase 1. Bioorg Med Chem 2017; 25:1622-1629. [PMID: 28139279 DOI: 10.1016/j.bmc.2017.01.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 12/15/2016] [Accepted: 01/16/2017] [Indexed: 10/20/2022]
Abstract
The first total synthesis of the antimicrobial natural product lynamicin D has been developed using a Suzuki coupling to construct the bisindole pyrrole skeleton. An evaluation of the biological activity of lynamicin D reveals that it has a minor effect on cell viability but it can modulate splicing of pre-mRNAs. We provide evidence that this effect is mainly due to the ability of lynamicin D to alter the levels of SRPK1, the key kinase involved in both constitutive and alternative splicing.
Collapse
Affiliation(s)
- Ioanna Sigala
- Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - George Ganidis
- Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Savvas Thysiadis
- Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Alexandros L Zografos
- Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Thomas Giannakouros
- Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Vasiliki Sarli
- Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece.
| | - Eleni Nikolakaki
- Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| |
Collapse
|
307
|
Eubanks CS, Forte JE, Kapral GJ, Hargrove AE. Small Molecule-Based Pattern Recognition To Classify RNA Structure. J Am Chem Soc 2017; 139:409-416. [PMID: 28004925 PMCID: PMC5465965 DOI: 10.1021/jacs.6b11087] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Three-dimensional RNA structures are notoriously difficult to determine, and the link between secondary structure and RNA conformation is only beginning to be understood. These challenges have hindered the identification of guiding principles for small molecule:RNA recognition. We herein demonstrate that the strong and differential binding ability of aminoglycosides to RNA structures can be used to classify five canonical RNA secondary structure motifs through principal component analysis (PCA). In these analyses, the aminoglycosides act as receptors, while RNA structures labeled with a benzofuranyluridine fluorophore act as analytes. Complete (100%) predictive ability for this RNA training set was achieved by incorporating two exhaustively guanidinylated aminoglycosides into the receptor library. The PCA was then externally validated using biologically relevant RNA constructs. In bulge-stem-loop constructs of HIV-1 transactivation response element (TAR) RNA, we achieved nucleotide-specific classification of two independent secondary structure motifs. Furthermore, examination of cheminformatic parameters and PCA loading factors revealed trends in aminoglycoside:RNA recognition, including the importance of shape-based discrimination, and suggested the potential for size and sequence discrimination within RNA structural motifs. These studies present a new approach to classifying RNA structure and provide direct evidence that RNA topology, in addition to sequence, is critical for the molecular recognition of RNA.
Collapse
Affiliation(s)
- Christopher S Eubanks
- Department of Chemistry, Duke University , Durham, North Carolina 27708, United States
| | - Jordan E Forte
- Department of Chemistry, Duke University , Durham, North Carolina 27708, United States
| | - Gary J Kapral
- Department of Chemistry, Duke University , Durham, North Carolina 27708, United States
| | - Amanda E Hargrove
- Department of Chemistry, Duke University , Durham, North Carolina 27708, United States
| |
Collapse
|
308
|
Approaches for the Discovery of Small Molecule Ligands Targeting microRNAs. TOPICS IN MEDICINAL CHEMISTRY 2017. [DOI: 10.1007/7355_2017_3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
309
|
Abstract
More than 95% of genes in the human genome are alternatively spliced to form multiple transcripts, often encoding proteins with differing or opposing function. The control of alternative splicing is now being elucidated, and with this comes the opportunity to develop modulators of alternative splicing that can control cellular function. A number of approaches have been taken to develop compounds that can experimentally, and sometimes clinically, affect splicing control, resulting in potential novel therapeutics. Here we develop the concepts that targeting alternative splicing can result in relatively specific pathway inhibitors/activators that result in dampening down of physiologic or pathologic processes, from changes in muscle physiology to altering angiogenesis or pain. The targets and pharmacology of some of the current inhibitors/activators of alternative splicing are demonstrated and future directions discussed.
Collapse
Affiliation(s)
- David O Bates
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (D.O.B.); School of Chemistry, UNSW Australia, Sydney, Australia (J.C.M.); School of Physiology, Pharmacology and Neurosciences, School of Clinical Sciences/Bristol Renal, University of Bristol, Bristol, United Kingdom (S.O.); and School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (L.F.D.)
| | - Jonathan C Morris
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (D.O.B.); School of Chemistry, UNSW Australia, Sydney, Australia (J.C.M.); School of Physiology, Pharmacology and Neurosciences, School of Clinical Sciences/Bristol Renal, University of Bristol, Bristol, United Kingdom (S.O.); and School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (L.F.D.)
| | - Sebastian Oltean
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (D.O.B.); School of Chemistry, UNSW Australia, Sydney, Australia (J.C.M.); School of Physiology, Pharmacology and Neurosciences, School of Clinical Sciences/Bristol Renal, University of Bristol, Bristol, United Kingdom (S.O.); and School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (L.F.D.)
| | - Lucy F Donaldson
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (D.O.B.); School of Chemistry, UNSW Australia, Sydney, Australia (J.C.M.); School of Physiology, Pharmacology and Neurosciences, School of Clinical Sciences/Bristol Renal, University of Bristol, Bristol, United Kingdom (S.O.); and School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (L.F.D.)
| |
Collapse
|
310
|
d'Ydewalle C, Ramos DM, Pyles NJ, Ng SY, Gorz M, Pilato CM, Ling K, Kong L, Ward AJ, Rubin LL, Rigo F, Bennett CF, Sumner CJ. The Antisense Transcript SMN-AS1 Regulates SMN Expression and Is a Novel Therapeutic Target for Spinal Muscular Atrophy. Neuron 2016; 93:66-79. [PMID: 28017471 DOI: 10.1016/j.neuron.2016.11.033] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 10/11/2016] [Accepted: 11/14/2016] [Indexed: 12/14/2022]
Abstract
The neuromuscular disorder spinal muscular atrophy (SMA), the most common inherited killer of infants, is caused by insufficient expression of survival motor neuron (SMN) protein. SMA therapeutics development efforts have focused on identifying strategies to increase SMN expression. We identified a long non-coding RNA (lncRNA) that arises from the antisense strand of SMN, SMN-AS1, which is enriched in neurons and transcriptionally represses SMN expression by recruiting the epigenetic Polycomb repressive complex-2. Targeted degradation of SMN-AS1 with antisense oligonucleotides (ASOs) increases SMN expression in patient-derived cells, cultured neurons, and the mouse central nervous system. SMN-AS1 ASOs delivered together with SMN2 splice-switching oligonucleotides additively increase SMN expression and improve survival of severe SMA mice. This study is the first proof of concept that targeting a lncRNA to transcriptionally activate SMN2 can be combined with SMN2 splicing modification to ameliorate SMA and demonstrates the promise of combinatorial ASOs for the treatment of neurogenetic disorders.
Collapse
Affiliation(s)
- Constantin d'Ydewalle
- Department of Neurology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Daniel M Ramos
- Department of Neuroscience, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Noah J Pyles
- Department of Neurology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Shi-Yan Ng
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Mariusz Gorz
- Department of Neurology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Celeste M Pilato
- Department of Neurology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Karen Ling
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - Lingling Kong
- Department of Neurology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Amanda J Ward
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02142, USA
| | - Lee L Rubin
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Frank Rigo
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - C Frank Bennett
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - Charlotte J Sumner
- Department of Neurology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA.
| |
Collapse
|
311
|
Disney MD, Angelbello AJ. Rational Design of Small Molecules Targeting Oncogenic Noncoding RNAs from Sequence. Acc Chem Res 2016; 49:2698-2704. [PMID: 27993012 DOI: 10.1021/acs.accounts.6b00326] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The discovery of RNA catalysis in the 1980s and the dissemination of the human genome sequence at the start of this century inspired investigations of the regulatory roles of noncoding RNAs in biology. In fact, the Encyclopedia of DNA Elements (ENCODE) project has shown that only 1-2% of the human genome encodes protein, yet 75% is transcribed into RNA. Functional studies both preceding and following the ENCODE project have shown that these noncoding RNAs have important roles in regulating gene expression, developmental timing, and other critical functions. RNA's diverse roles are often a consequence of the various folds that it adopts. The single-stranded nature of the biopolymer enables it to adopt intramolecular folds with noncanonical pairings to lower its free energy. These folds can be scaffolds to bind proteins or to form frameworks to interact with other RNAs. Not surprisingly, dysregulation of certain noncoding RNAs has been shown to be causative of disease. Given this as the background, it is easy to see why it would be useful to develop methods that target RNA and manipulate its biology in rational and predictable ways. The antisense approach has afforded strategies to target RNAs via Watson-Crick base pairing and has typically focused on targeting partially unstructured regions of RNA. Small molecule strategies to target RNA would be desirable not only because compounds could be lead optimized via medicinal chemistry but also because structured regions within an RNA of interest could be targeted to directly interfere with RNA folds that contribute to disease. Additionally, small molecules have historically been the most successful drug candidates. Until recently, the ability to design small molecules that target non-ribosomal RNAs has been elusive, creating the perception that they are "undruggable". In this Account, approaches to demystify targeting RNA with small molecules are described. Rather than bulk screening for compounds that bind to singular targets, which is the purview of the pharmaceutical industry and academic institutions with high throughput screening facilities, we focus on methods that allow for the rational design of small molecules toward biological RNAs. One enabling and foundational technology that has been developed is two-dimensional combinatorial screening (2DCS), a library-versus-library selection approach that allows the identification of the RNA motif binding preferences of small molecules from millions of combinations. A landscape map of the 2DCS-defined and annotated RNA motif-small molecule interactions is then placed into Inforna, a computational tool that allows one to mine these interactions against an RNA of interest or an entire transcriptome. Indeed, this approach has been enabled by tools to annotate RNA structure from sequence, an invaluable asset to the RNA community and this work, and has allowed for the rational identification of "druggable" RNAs in a target agnostic fashion.
Collapse
Affiliation(s)
- Matthew D. Disney
- Departments of
Chemistry
and Neuroscience, The Scripps Research Institute, 130 Scripps Way, #3A1, Jupiter, Florida 33458, United States
| | - Alicia J. Angelbello
- Departments of
Chemistry
and Neuroscience, The Scripps Research Institute, 130 Scripps Way, #3A1, Jupiter, Florida 33458, United States
| |
Collapse
|
312
|
Abstract
Defined genetic models based on human pluripotent stem cells have opened new avenues for understanding disease mechanisms and drug screening. Many of these models assume cell-autonomous mechanisms of disease but it is possible that disease phenotypes or drug responses will only be evident if all cellular and extracellular components of a tissue are present and functionally mature. To derive optimal benefit from such models, complex multicellular structures with vascular components that mimic tissue niches will thus likely be necessary. Here we consider emerging research creating human tissue mimics and provide some recommendations for moving the field forward.
Collapse
|
313
|
Arnold WD, Duque S, Iyer CC, Zaworski P, McGovern VL, Taylor SJ, von Herrmann KM, Kobayashi DT, Chen KS, Kolb SJ, Paushkin SV, Burghes AHM. Normalization of Patient-Identified Plasma Biomarkers in SMNΔ7 Mice following Postnatal SMN Restoration. PLoS One 2016; 11:e0167077. [PMID: 27907033 PMCID: PMC5132001 DOI: 10.1371/journal.pone.0167077] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 11/08/2016] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION AND OBJECTIVE Spinal muscular atrophy (SMA) is an autosomal recessive motor neuron disorder. SMA is caused by homozygous loss of the SMN1 gene and retention of the SMN2 gene resulting in reduced levels of full length SMN protein that are insufficient for motor neuron function. Various treatments that restore levels of SMN are currently in clinical trials and biomarkers are needed to determine the response to treatment. Here, we sought to investigate in SMA mice a set of plasma analytes, previously identified in patients with SMA to correlate with motor function. The goal was to determine whether levels of plasma markers were altered in the SMNΔ7 mouse model of SMA and whether postnatal SMN restoration resulted in normalization of the biomarkers. METHODS SMNΔ7 and control mice were treated with antisense oligonucleotides (ASO) targeting ISS-N1 to increase SMN protein from SMN2 or scramble ASO (sham treatment) via intracerebroventricular injection on postnatal day 1 (P1). Brain, spinal cord, quadriceps muscle, and liver were analyzed for SMN protein levels at P12 and P90. Ten plasma biomarkers (a subset of biomarkers in the SMA-MAP panel available for analysis in mice) were analyzed in plasma obtained at P12, P30, and P90. RESULTS Of the eight plasma biomarkers assessed, 5 were significantly changed in sham treated SMNΔ7 mice compared to control mice and were normalized in SMNΔ7 mice treated with ASO. CONCLUSION This study defines a subset of the SMA-MAP plasma biomarker panel that is abnormal in the most commonly used mouse model of SMA. Furthermore, some of these markers are responsive to postnatal SMN restoration. These findings support continued clinical development of these potential prognostic and pharmacodynamic biomarkers.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Biomarkers/metabolism
- Brain/metabolism
- Brain/pathology
- Clinical Trials as Topic
- Disease Models, Animal
- Gene Expression Regulation
- Genetic Complementation Test
- Humans
- Injections, Intraventricular
- Liver/metabolism
- Liver/pathology
- Mice
- Mice, Transgenic
- Motor Neurons/metabolism
- Motor Neurons/pathology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Atrophy, Spinal/genetics
- Muscular Atrophy, Spinal/metabolism
- Muscular Atrophy, Spinal/pathology
- Oligonucleotides, Antisense/genetics
- Oligonucleotides, Antisense/metabolism
- Spinal Cord/metabolism
- Spinal Cord/pathology
- Survival of Motor Neuron 1 Protein/genetics
- Survival of Motor Neuron 1 Protein/metabolism
- Survival of Motor Neuron 2 Protein/genetics
- Survival of Motor Neuron 2 Protein/metabolism
Collapse
Affiliation(s)
- W. David Arnold
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus Ohio, United States of America
- Department of Physical Medicine and Rehabilitation, The Ohio State University Wexner Medical Center, Columbus Ohio, United States of America
| | - Sandra Duque
- VIB Center for the Biology of Disease – KU Leuven Department of Human Genetics, Leuven Belgium, United States of America
| | - Chitra C. Iyer
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, Columbus Ohio, United States of America
| | | | - Vicki L. McGovern
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, Columbus Ohio, United States of America
| | | | | | | | - Karen S. Chen
- SMA Foundation, New York, New York, United States of America
| | - Stephen J. Kolb
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus Ohio, United States of America
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, Columbus Ohio, United States of America
| | | | - Arthur H. M. Burghes
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus Ohio, United States of America
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, Columbus Ohio, United States of America
- * E-mail:
| |
Collapse
|
314
|
Calder AN, Androphy EJ, Hodgetts KJ. Small Molecules in Development for the Treatment of Spinal Muscular Atrophy. J Med Chem 2016; 59:10067-10083. [PMID: 27490705 PMCID: PMC5744254 DOI: 10.1021/acs.jmedchem.6b00670] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive neurodegenerative disease resulting from pathologically low levels of survival motor neuron (SMN) protein. The majority of mRNA from the SMN2 allele undergoes alternative splicing and excludes critical codons, causing an SMN protein deficiency. While there is currently no FDA-approved treatment for SMA, early therapeutic efforts have focused on testing repurposed drugs such as phenylbutyrate (2), valproic acid (3), riluzole (6), hydroxyurea (7), and albuterol (9), none of which has demonstrated clinical effectiveness. More recently, clinical trials have focused on novel small-molecule compounds identified from high-throughput screening and medicinal chemistry optimization such as olesoxime (11), CK-2127107, RG7800, LMI070, and RG3039 (17). In this paper, we review both repurposed drugs and small-molecule compounds discovered following medicinal chemistry optimization for the potential treatment of SMA.
Collapse
Affiliation(s)
- Alyssa N. Calder
- Laboratory for Drug Discovery in Neurodegeneration, Brigham & Women’s Hospital and Harvard Medical School, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - Elliot J. Androphy
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kevin J. Hodgetts
- Laboratory for Drug Discovery in Neurodegeneration, Brigham & Women’s Hospital and Harvard Medical School, 65 Landsdowne Street, Cambridge, MA 02139, USA
| |
Collapse
|
315
|
Abera MB, Xiao J, Nofziger J, Titus S, Southall N, Zheng W, Moritz KE, Ferrer M, Cherry JJ, Androphy EJ, Wang A, Xu X, Austin C, Fischbeck KH, Marugan JJ, Burnett BG. ML372 blocks SMN ubiquitination and improves spinal muscular atrophy pathology in mice. JCI Insight 2016; 1:e88427. [PMID: 27882347 DOI: 10.1172/jci.insight.88427] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive neuromuscular disease and one of the leading inherited causes of infant mortality. SMA results from insufficient levels of the survival motor neuron (SMN) protein, and studies in animal models of the disease have shown that increasing SMN protein levels ameliorates the disease phenotype. Our group previously identified and optimized a new series of small molecules, with good potency and toxicity profiles and reasonable pharmacokinetics, that were able to increase SMN protein levels in SMA patient-derived cells. We show here that ML372, a representative of this series, almost doubles the half-life of residual SMN protein expressed from the SMN2 locus by blocking its ubiquitination and subsequent degradation by the proteasome. ML372 increased SMN protein levels in muscle, spinal cord, and brain tissue of SMA mice. Importantly, ML372 treatment improved the righting reflex and extended survival of a severe mouse model of SMA. These results demonstrate that slowing SMN degradation by selectively inhibiting its ubiquitination can improve the motor phenotype and lifespan of SMA model mice.
Collapse
Affiliation(s)
- Mahlet B Abera
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hebert School of Medicine, Bethesda, Maryland, USA
| | - Jingbo Xiao
- NIH Chemical Genomics Center, Discovery Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Jonathan Nofziger
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Steve Titus
- NIH Chemical Genomics Center, Discovery Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Noel Southall
- NIH Chemical Genomics Center, Discovery Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Wei Zheng
- NIH Chemical Genomics Center, Discovery Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Kasey E Moritz
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hebert School of Medicine, Bethesda, Maryland, USA
| | - Marc Ferrer
- NIH Chemical Genomics Center, Discovery Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Jonathan J Cherry
- Department of Dermatology,, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Elliot J Androphy
- Department of Dermatology,, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Amy Wang
- NIH Chemical Genomics Center, Discovery Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Xin Xu
- NIH Chemical Genomics Center, Discovery Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Christopher Austin
- NIH Chemical Genomics Center, Discovery Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Kenneth H Fischbeck
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Juan J Marugan
- NIH Chemical Genomics Center, Discovery Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Barrington G Burnett
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hebert School of Medicine, Bethesda, Maryland, USA
| |
Collapse
|
316
|
Fletcher S, Bellgard MI, Price L, Akkari AP, Wilton SD. Translational development of splice-modifying antisense oligomers. Expert Opin Biol Ther 2016; 17:15-30. [PMID: 27805416 DOI: 10.1080/14712598.2017.1250880] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Antisense nucleic acid analogues can interact with pre-mRNA motifs and influence exon or splice site selection and thereby alter gene expression. Design of antisense molecules to target specific motifs can result in either exon exclusion or exon inclusion during splicing. Novel drugs exploiting the antisense concept are targeting rare, life-limiting diseases; however, the potential exists to treat a wide range of conditions by antisense-mediated splice intervention. Areas covered: In this review, the authors discuss the clinical translation of novel molecular therapeutics to address the fatal neuromuscular disorders Duchenne muscular dystrophy and spinal muscular atrophy. The review also highlights difficulties posed by issues pertaining to restricted participant numbers, variable phenotype and disease progression, and the identification and validation of study endpoints. Expert opinion: Translation of novel therapeutics for Duchenne muscular dystrophy and spinal muscular atrophy has been greatly advanced by multidisciplinary research, academic-industry partnerships and in particular, the engagement and support of the patient community. Sponsors, supporters and regulators are cooperating to deliver new drugs and identify and define meaningful outcome measures. Non-conventional and adaptive trial design could be particularly suited to clinical evaluation of novel therapeutics and strategies to treat serious, rare diseases that may be problematic to study using more conventional clinical trial structures.
Collapse
Affiliation(s)
- S Fletcher
- a Centre for Neuromuscular and Neurological Disorders , University of Western Australia , Nedlands , Western Australia , Australia.,b Western Australian Neuroscience Research Institute , Nedlands , Western Australia , Australia.,c Centre for Comparative Genomics , Murdoch University , Western Australia , Australia
| | - M I Bellgard
- b Western Australian Neuroscience Research Institute , Nedlands , Western Australia , Australia.,c Centre for Comparative Genomics , Murdoch University , Western Australia , Australia
| | - L Price
- a Centre for Neuromuscular and Neurological Disorders , University of Western Australia , Nedlands , Western Australia , Australia.,b Western Australian Neuroscience Research Institute , Nedlands , Western Australia , Australia.,c Centre for Comparative Genomics , Murdoch University , Western Australia , Australia
| | - A P Akkari
- b Western Australian Neuroscience Research Institute , Nedlands , Western Australia , Australia.,c Centre for Comparative Genomics , Murdoch University , Western Australia , Australia.,d Shiraz Pharmaceuticals, Inc , Chapel Hill , NC , USA
| | - S D Wilton
- a Centre for Neuromuscular and Neurological Disorders , University of Western Australia , Nedlands , Western Australia , Australia.,b Western Australian Neuroscience Research Institute , Nedlands , Western Australia , Australia.,c Centre for Comparative Genomics , Murdoch University , Western Australia , Australia
| |
Collapse
|
317
|
Garber K. Big win possible for Ionis/Biogen antisense drug in muscular atrophy. Nat Biotechnol 2016; 34:1002-1003. [PMID: 27727217 DOI: 10.1038/nbt1016-1002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
318
|
Armbruster N, Lattanzi A, Jeavons M, Van Wittenberghe L, Gjata B, Marais T, Martin S, Vignaud A, Voit T, Mavilio F, Barkats M, Buj-Bello A. Efficacy and biodistribution analysis of intracerebroventricular administration of an optimized scAAV9-SMN1 vector in a mouse model of spinal muscular atrophy. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:16060. [PMID: 27652289 PMCID: PMC5022869 DOI: 10.1038/mtm.2016.60] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/12/2016] [Accepted: 07/15/2016] [Indexed: 12/13/2022]
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive disease of variable severity caused by mutations in the SMN1 gene. Deficiency of the ubiquitous SMN function results in spinal cord α-motor neuron degeneration and proximal muscle weakness. Gene replacement therapy with recombinant adeno-associated viral (AAV) vectors showed therapeutic efficacy in several animal models of SMA. Here, we report a study aimed at analyzing the efficacy and biodistribution of a serotype-9, self-complementary AAV vector expressing a codon-optimized human SMN1 coding sequence (coSMN1) under the control of the constitutive phosphoglycerate kinase (PGK) promoter in neonatal SMNΔ7 mice, a severe animal model of the disease. We administered the scAAV9-coSMN1 vector in the intracerebroventricular (ICV) space in a dose-escalating mode, and analyzed survival, vector biodistribution and SMN protein expression in the spinal cord and peripheral tissues. All treated mice showed a significant, dose-dependent rescue of lifespan and growth with a median survival of 346 days. Additional administration of vector by an intravenous route (ICV+IV) did not improve survival, and vector biodistribution analysis 90 days postinjection indicated that diffusion from the cerebrospinal fluid to the periphery was sufficient to rescue the SMA phenotype. These results support the preclinical development of SMN1 gene therapy by CSF vector delivery.
Collapse
Affiliation(s)
| | | | | | | | | | - Thibaut Marais
- Center of Research in Myology, INSERM UMRS 974, CNRS FRE 3617, Institut de Myologie, Université Pierre et Marie Curie Paris 6 , Paris, France
| | | | | | - Thomas Voit
- Center of Research in Myology, INSERM UMRS 974, CNRS FRE 3617, Institut de Myologie, Université Pierre et Marie Curie Paris 6 , Paris, France
| | | | - Martine Barkats
- Center of Research in Myology, INSERM UMRS 974, CNRS FRE 3617, Institut de Myologie, Université Pierre et Marie Curie Paris 6 , Paris, France
| | - Ana Buj-Bello
- INSERM UMR 951, Evry, France; Genethon, Evry, France
| |
Collapse
|
319
|
Abstract
Tumor-associated alterations in RNA splicing result either from mutations in splicing-regulatory elements or changes in components of the splicing machinery. This review summarizes our current understanding of the role of splicing-factor alterations in human cancers. We describe splicing-factor alterations detected in human tumors and the resulting changes in splicing, highlighting cell-type-specific similarities and differences. We review the mechanisms of splicing-factor regulation in normal and cancer cells. Finally, we summarize recent efforts to develop novel cancer therapies, based on targeting either the oncogenic splicing events or their upstream splicing regulators.
Collapse
Affiliation(s)
- Olga Anczuków
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Adrian R Krainer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| |
Collapse
|
320
|
Tseng YT, Chen CS, Jong YJ, Chang FR, Lo YC. Loganin possesses neuroprotective properties, restores SMN protein and activates protein synthesis positive regulator Akt/mTOR in experimental models of spinal muscular atrophy. Pharmacol Res 2016; 111:58-75. [DOI: 10.1016/j.phrs.2016.05.023] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 05/03/2016] [Accepted: 05/04/2016] [Indexed: 12/21/2022]
|
321
|
Connelly CM, Moon MH, Schneekloth JS. The Emerging Role of RNA as a Therapeutic Target for Small Molecules. Cell Chem Biol 2016; 23:1077-1090. [PMID: 27593111 PMCID: PMC5064864 DOI: 10.1016/j.chembiol.2016.05.021] [Citation(s) in RCA: 231] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/07/2016] [Accepted: 05/18/2016] [Indexed: 01/09/2023]
Abstract
Recent advances in understanding different RNAs and unique features of their biology have revealed a wealth of information. However, approaches to identify small molecules that target these newly discovered regulatory elements have been lacking. The application of new biochemical screening and design-based technologies, coupled with a resurgence of interest in phenotypic screening, has resulted in several compelling successes in targeting RNA. A number of recent advances suggest that achieving the long-standing goal of developing drug-like, biologically active small molecules that target RNA is possible. This review highlights advances and successes in approaches to targeting RNA with diverse small molecules, and the potential for these technologies to pave the way to new types of RNA-targeted therapeutics.
Collapse
Affiliation(s)
- Colleen M Connelly
- Chemical Biology Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | - Michelle H Moon
- Chemical Biology Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | - John S Schneekloth
- Chemical Biology Laboratory, National Cancer Institute, Frederick, MD 21702, USA.
| |
Collapse
|
322
|
Somatic Therapy of a Mouse SMA Model with a U7 snRNA Gene Correcting SMN2 Splicing. Mol Ther 2016; 24:1797-1805. [PMID: 27456062 PMCID: PMC5112044 DOI: 10.1038/mt.2016.152] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 07/18/2016] [Indexed: 12/12/2022] Open
Abstract
Spinal Muscular Atrophy is due to the loss of SMN1 gene function. The duplicate gene SMN2 produces some, but not enough, SMN protein because most transcripts lack exon 7. Thus, promoting the inclusion of this exon is a therapeutic option. We show that a somatic gene therapy using the gene for a modified U7 RNA which stimulates this splicing has a profound and persistent therapeutic effect on the phenotype of a severe Spinal Muscular Atrophy mouse model. To this end, the U7 gene and vector and the production of pure, highly concentrated self-complementary (sc) adenovirus-associated virus 9 vector particles were optimized. Introduction of the functional vector into motoneurons of newborn Spinal Muscular Atrophy mice by intracerebroventricular injection led to a highly significant, dose-dependent increase in life span and improvement of muscle functions. Besides the central nervous system, the therapeutic U7 RNA was expressed in the heart and liver which may additionally have contributed to the observed therapeutic efficacy. This approach provides an additional therapeutic option for Spinal Muscular Atrophy and could also be adapted to treat other diseases of the central nervous system with regulatory small RNA genes.
Collapse
|
323
|
Osman EY, Washington CW, Kaifer KA, Mazzasette C, Patitucci TN, Florea KM, Simon ME, Ko CP, Ebert AD, Lorson CL. Optimization of Morpholino Antisense Oligonucleotides Targeting the Intronic Repressor Element1 in Spinal Muscular Atrophy. Mol Ther 2016; 24:1592-601. [PMID: 27401142 DOI: 10.1038/mt.2016.145] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 07/04/2016] [Indexed: 12/13/2022] Open
Abstract
Loss of Survival Motor Neuron-1 (SMN1) causes Spinal Muscular Atrophy, a devastating neurodegenerative disease. SMN2 is a nearly identical copy gene; however SMN2 cannot prevent disease development in the absence of SMN1 since the majority of SMN2-derived transcripts are alternatively spliced, encoding a truncated, unstable protein lacking exon 7. Nevertheless, SMN2 retains the ability to produce low levels of functional protein. Previously we have described a splice-switching Morpholino antisense oligonucleotide (ASO) sequence that targets a potent intronic repressor, Element1 (E1), located upstream of SMN2 exon 7. In this study, we have assessed a novel panel of Morpholino ASOs with the goal of optimizing E1 ASO activity. Screening for efficacy in the SMNΔ7 mouse model, a single ASO variant was more active in vivo compared with the original E1(MO)-ASO. Sequence variant eleven (E1(MOv11)) consistently showed greater efficacy by increasing the lifespan of severe Spinal Muscular Atrophy mice after a single intracerebroventricular injection in the central nervous system, exhibited a strong dose-response across an order of magnitude, and demonstrated excellent target engagement by partially reversing the pathogenic SMN2 splicing event. We conclude that Morpholino modified ASOs are effective in modifying SMN2 splicing and have the potential for future Spinal Muscular Atrophy clinical applications.
Collapse
Affiliation(s)
- Erkan Y Osman
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
| | - Charles W Washington
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
| | - Kevin A Kaifer
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA.,Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Chiara Mazzasette
- Department of Biological Sciences, University of Southern California, Los Angeles, California, USA
| | - Teresa N Patitucci
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Kyra M Florea
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA.,Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Madeline E Simon
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA.,Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Chien-Ping Ko
- Department of Biological Sciences, University of Southern California, Los Angeles, California, USA
| | - Allison D Ebert
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Christian L Lorson
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA.,Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
324
|
Ratni H, Karp GM, Weetall M, Naryshkin NA, Paushkin SV, Chen KS, McCarthy KD, Qi H, Turpoff A, Woll MG, Zhang X, Zhang N, Yang T, Dakka A, Vazirani P, Zhao X, Pinard E, Green L, David-Pierson P, Tuerck D, Poirier A, Muster W, Kirchner S, Mueller L, Gerlach I, Metzger F. Specific Correction of Alternative Survival Motor Neuron 2 Splicing by Small Molecules: Discovery of a Potential Novel Medicine To Treat Spinal Muscular Atrophy. J Med Chem 2016; 59:6086-100. [DOI: 10.1021/acs.jmedchem.6b00459] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Hasane Ratni
- pRED, Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Gary M. Karp
- PTC Therapeutics, Inc., 100
Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Marla Weetall
- PTC Therapeutics, Inc., 100
Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Nikolai A. Naryshkin
- PTC Therapeutics, Inc., 100
Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Sergey V. Paushkin
- SMA Foundation, 888 Seventh
Avenue, Suite 400, New York, New York 10019, United States
| | - Karen S. Chen
- SMA Foundation, 888 Seventh
Avenue, Suite 400, New York, New York 10019, United States
| | - Kathleen D. McCarthy
- SMA Foundation, 888 Seventh
Avenue, Suite 400, New York, New York 10019, United States
| | - Hongyan Qi
- PTC Therapeutics, Inc., 100
Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Anthony Turpoff
- PTC Therapeutics, Inc., 100
Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Matthew G. Woll
- PTC Therapeutics, Inc., 100
Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Xiaoyan Zhang
- PTC Therapeutics, Inc., 100
Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Nanjing Zhang
- PTC Therapeutics, Inc., 100
Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Tianle Yang
- PTC Therapeutics, Inc., 100
Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Amal Dakka
- PTC Therapeutics, Inc., 100
Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Priya Vazirani
- PTC Therapeutics, Inc., 100
Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Xin Zhao
- PTC Therapeutics, Inc., 100
Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Emmanuel Pinard
- pRED, Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Luke Green
- pRED, Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Pascale David-Pierson
- pRED, Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Dietrich Tuerck
- pRED, Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Agnes Poirier
- pRED, Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Wolfgang Muster
- pRED, Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Stephan Kirchner
- pRED, Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Lutz Mueller
- pRED, Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Irene Gerlach
- pRED, Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Friedrich Metzger
- pRED, Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland
| |
Collapse
|
325
|
Woll MG, Qi H, Turpoff A, Zhang N, Zhang X, Chen G, Li C, Huang S, Yang T, Moon YC, Lee CS, Choi S, Almstead NG, Naryshkin NA, Dakka A, Narasimhan J, Gabbeta V, Welch E, Zhao X, Risher N, Sheedy J, Weetall M, Karp GM. Discovery and Optimization of Small Molecule Splicing Modifiers of Survival Motor Neuron 2 as a Treatment for Spinal Muscular Atrophy. J Med Chem 2016; 59:6070-85. [DOI: 10.1021/acs.jmedchem.6b00460] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Matthew G. Woll
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Hongyan Qi
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Anthony Turpoff
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Nanjing Zhang
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Xiaoyan Zhang
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Guangming Chen
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Chunshi Li
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Song Huang
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Tianle Yang
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Young-Choon Moon
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Chang-Sun Lee
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Soongyu Choi
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Neil G. Almstead
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Nikolai A. Naryshkin
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Amal Dakka
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Jana Narasimhan
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Vijayalakshmi Gabbeta
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Ellen Welch
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Xin Zhao
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Nicole Risher
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Josephine Sheedy
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Marla Weetall
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| | - Gary M. Karp
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, New Jersey 07080, United States
| |
Collapse
|
326
|
Catapano F, Zaharieva I, Scoto M, Marrosu E, Morgan J, Muntoni F, Zhou H. Altered Levels of MicroRNA-9, -206, and -132 in Spinal Muscular Atrophy and Their Response to Antisense Oligonucleotide Therapy. MOLECULAR THERAPY. NUCLEIC ACIDS 2016; 5:e331. [PMID: 27377135 PMCID: PMC5014531 DOI: 10.1038/mtna.2016.47] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 05/25/2016] [Indexed: 12/12/2022]
Abstract
The identification of noninvasive biomarkers to monitor the disease progression in spinal muscular atrophy (SMA) is becoming increasingly important. MicroRNAs (miRNAs) regulate gene expression and are implicated in the pathogenesis of neuromuscular diseases, including motor neuron degeneration. In this study, we selectively characterized the expression of miR-9, miR-206, and miR-132 in spinal cord, skeletal muscle, and serum from SMA transgenic mice, and in serum from SMA patients. A systematic analysis of miRNA expression was conducted in SMA mice with different disease severities (severe type I-like and mild type III-like) at different disease stages (pre-, mid-, and late-symptomatic stages), and in morpholino antisense oligonucleotide-treated mice. There was differential expression of all three miRNAs in spinal cord, skeletal muscle and serum samples in SMA mice. Serum miRNAs were altered prior to the changes in spinal cord and skeletal muscle at the presymptomatic stage. The altered miR-132 levels in spinal cord, muscle, and serum transiently reversed to normal level after a single-dose morpholino antisense oligomer PMO25 treatment in SMA mice. We also confirmed a significant alteration of miR-9 and miR-132 level in serum samples from SMA patients. Our study indicates the potential of developing miRNAs as noninvasive biomarkers in SMA.
Collapse
Affiliation(s)
- Francesco Catapano
- Dubowitz Neuromuscular Centre and Developmental Neuroscience Programme, Institute of Child Health, University College London, London, UK
| | - Irina Zaharieva
- Dubowitz Neuromuscular Centre and Developmental Neuroscience Programme, Institute of Child Health, University College London, London, UK
| | - Mariacristina Scoto
- Dubowitz Neuromuscular Centre and Developmental Neuroscience Programme, Institute of Child Health, University College London, London, UK
| | - Elena Marrosu
- Dubowitz Neuromuscular Centre and Developmental Neuroscience Programme, Institute of Child Health, University College London, London, UK
| | - Jennifer Morgan
- Dubowitz Neuromuscular Centre and Developmental Neuroscience Programme, Institute of Child Health, University College London, London, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre and Developmental Neuroscience Programme, Institute of Child Health, University College London, London, UK
| | - Haiyan Zhou
- Dubowitz Neuromuscular Centre and Developmental Neuroscience Programme, Institute of Child Health, University College London, London, UK
| |
Collapse
|
327
|
Tudor M, Hermes J, Li J. Translatability: what does it mean in drug discovery? Drug Discov Today 2016; 21:865-7. [DOI: 10.1016/j.drudis.2016.03.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 03/02/2016] [Accepted: 03/18/2016] [Indexed: 12/21/2022]
|
328
|
Molecular Basis and Therapeutic Strategies to Rescue Factor IX Variants That Affect Splicing and Protein Function. PLoS Genet 2016; 12:e1006082. [PMID: 27227676 PMCID: PMC4882169 DOI: 10.1371/journal.pgen.1006082] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/04/2016] [Indexed: 12/24/2022] Open
Abstract
Mutations that result in amino acid changes can affect both pre-mRNA splicing and protein function. Understanding the combined effect is essential for correct diagnosis and for establishing the most appropriate therapeutic strategy at the molecular level. We have identified a series of disease-causing splicing mutations in coagulation factor IX (FIX) exon 5 that are completely recovered by a modified U1snRNP particle, through an SRSF2-dependent enhancement mechanism. We discovered that synonymous mutations and missense substitutions associated to a partial FIX secretion defect represent targets for this therapy as the resulting spliced-corrected proteins maintains normal FIX coagulant specific activity. Thus, splicing and protein alterations contribute to define at the molecular level the disease-causing effect of a number of exonic mutations in coagulation FIX exon 5. In addition, our results have a significant impact in the development of splicing-switching therapies in particular for mutations that affect both splicing and protein function where increasing the amount of a correctly spliced protein can circumvent the basic functional defects.
Collapse
|
329
|
De Vos J, Bouckenheimer J, Sansac C, Lemaître JM, Assou S. Human induced pluripotent stem cells: A disruptive innovation. Curr Res Transl Med 2016; 64:91-6. [PMID: 27316392 DOI: 10.1016/j.retram.2016.04.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/04/2016] [Accepted: 04/08/2016] [Indexed: 12/31/2022]
Abstract
This year (2016) will mark the 10th anniversary of the discovery of induced pluripotent stem cells (iPSCs). The finding that the transient expression of four transcription factors can radically remodel the epigenome, transcriptome and metabolome of differentiated cells and reprogram them into pluripotent stem cells has been a major and groundbreaking technological innovation. In this review, we discuss the major applications of this technology that we have grouped in nine categories: a model to study cell fate control; a model to study pluripotency; a model to study human development; a model to study human tissue and organ physiology; a model to study genetic diseases in a dish; a tool for cell rejuvenation; a source of cells for drug screening; a source of cells for regenerative medicine; a tool for the production of human organs in animals.
Collapse
Affiliation(s)
- J De Vos
- CHU Montpellier, Institute for Regenerative Medicine and Biotherapy, Hôpital Saint-Eloi, 34000 Montpellier, France; INSERM, U1183, 34000 Montpellier, France; Université de Montpellier, UFR de Médecine, 34000 Montpellier, France; Institut de Biologie Computationnelle, 34000 Montpellier, France; CHU Montpellier, SAFE-IPS Reprogramming Platform, Institute of Research in Biotherapy, 34000 Montpellier, France; CHU Montpellier, Unit for Cellular Therapy, Hospital Saint-Eloi, 34000 Montpellier, France.
| | - J Bouckenheimer
- CHU Montpellier, Institute for Regenerative Medicine and Biotherapy, Hôpital Saint-Eloi, 34000 Montpellier, France; INSERM, U1183, 34000 Montpellier, France; Université de Montpellier, UFR de Pharmacie, 34000 Montpellier, France
| | - C Sansac
- CHU Montpellier, Institute for Regenerative Medicine and Biotherapy, Hôpital Saint-Eloi, 34000 Montpellier, France; INSERM, U1183, 34000 Montpellier, France; Université de Montpellier, UFR de Pharmacie, 34000 Montpellier, France
| | - J-M Lemaître
- CHU Montpellier, Institute for Regenerative Medicine and Biotherapy, Hôpital Saint-Eloi, 34000 Montpellier, France; INSERM, U1183, 34000 Montpellier, France
| | - S Assou
- CHU Montpellier, Institute for Regenerative Medicine and Biotherapy, Hôpital Saint-Eloi, 34000 Montpellier, France; INSERM, U1183, 34000 Montpellier, France; Université de Montpellier, UFR de Médecine, 34000 Montpellier, France.
| |
Collapse
|
330
|
Abstract
Examples of associations between human disease and defects in pre-messenger RNA splicing/alternative splicing are accumulating. Although many alterations are caused by mutations in splicing signals or regulatory sequence elements, recent studies have noted the disruptive impact of mutated generic spliceosome components and splicing regulatory proteins. This review highlights recent progress in our understanding of how the altered splicing function of RNA-binding proteins contributes to myelodysplastic syndromes, cancer, and neuropathologies.
Collapse
Affiliation(s)
- Benoit Chabot
- Centre of Excellence in RNA Biology, Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec J1E 4K8, Canada
| | - Lulzim Shkreta
- Centre of Excellence in RNA Biology, Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec J1E 4K8, Canada
| |
Collapse
|
331
|
Sintusek P, Catapano F, Angkathunkayul N, Marrosu E, Parson SH, Morgan JE, Muntoni F, Zhou H. Histopathological Defects in Intestine in Severe Spinal Muscular Atrophy Mice Are Improved by Systemic Antisense Oligonucleotide Treatment. PLoS One 2016; 11:e0155032. [PMID: 27163330 PMCID: PMC4862622 DOI: 10.1371/journal.pone.0155032] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 04/22/2016] [Indexed: 12/28/2022] Open
Abstract
Gastrointestinal (GI) defects, including gastroesophageal reflux, constipation and delayed gastric emptying, are common in patients with spinal muscular atrophy (SMA). Similar GI dysmotility has been identified in mouse models with survival of motor neuron (SMN) protein deficiency. We previously described vascular defects in skeletal muscle and spinal cord of SMA mice and we hypothesized that similar defects could be involved in the GI pathology observed in these mice. We therefore investigated the gross anatomical structure, enteric vasculature and neurons in the small intestine in a severe mouse model of SMA. We also assessed the therapeutic response of GI histopathology to systemic administration of morpholino antisense oligonucleotide (AON) designed to increase SMN protein expression. Significant anatomical and histopathological abnormalities, with striking reduction of vascular density, overabundance of enteric neurons and increased macrophage infiltration, were detected in the small intestine in SMA mice. After systemic AON treatment in neonatal mice, all the abnormalities observed were significantly restored to near-normal levels. We conclude that the observed GI histopathological phenotypes and functional defects observed in these SMA mice are strongly linked to SMN deficiency which can be rescued by systemic administration of AON. This study on the histopathological changes in the gastrointestinal system in severe SMA mice provides further indication of the complex role that SMN plays in multiple tissues and suggests that at least in SMA mice restoration of SMN production in peripheral tissues is essential for optimal outcome.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Disease Models, Animal
- Gene Expression
- Genetic Therapy/methods
- Heterozygote
- Homozygote
- Humans
- Injections, Subcutaneous
- Intestine, Small/blood supply
- Intestine, Small/drug effects
- Intestine, Small/innervation
- Intestine, Small/pathology
- Macrophages/drug effects
- Macrophages/metabolism
- Macrophages/pathology
- Mice
- Mice, Transgenic
- Morpholinos/administration & dosage
- Motor Neurons/drug effects
- Motor Neurons/metabolism
- Motor Neurons/pathology
- Muscular Atrophy, Spinal/genetics
- Muscular Atrophy, Spinal/metabolism
- Muscular Atrophy, Spinal/pathology
- Muscular Atrophy, Spinal/therapy
- Oligonucleotides, Antisense/administration & dosage
- Phenotype
- Spinal Cord/blood supply
- Spinal Cord/drug effects
- Spinal Cord/pathology
- Survival of Motor Neuron 1 Protein/agonists
- Survival of Motor Neuron 1 Protein/genetics
- Survival of Motor Neuron 1 Protein/metabolism
Collapse
Affiliation(s)
- Palittiya Sintusek
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, London, United Kingdom
- Division of Gastroenterology and Hepatology, Department of Pediatrics, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, Thailand
| | - Francesco Catapano
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, London, United Kingdom
| | - Napat Angkathunkayul
- Department of Pathology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Elena Marrosu
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, London, United Kingdom
| | - Simon H. Parson
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
- Euan MacDonald Center for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Jennifer E. Morgan
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, London, United Kingdom
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, London, United Kingdom
- * E-mail: (HZ); (FM)
| | - Haiyan Zhou
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, London, United Kingdom
- * E-mail: (HZ); (FM)
| |
Collapse
|
332
|
Butchbach MER, Lumpkin CJ, Harris AW, Saieva L, Edwards JD, Workman E, Simard LR, Pellizzoni L, Burghes AHM. Protective effects of butyrate-based compounds on a mouse model for spinal muscular atrophy. Exp Neurol 2016; 279:13-26. [PMID: 26892876 PMCID: PMC4834225 DOI: 10.1016/j.expneurol.2016.02.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/11/2016] [Accepted: 02/13/2016] [Indexed: 11/17/2022]
Abstract
Proximal spinal muscular atrophy (SMA) is a childhood-onset degenerative disease resulting from the selective loss of motor neurons in the spinal cord. SMA is caused by the loss of SMN1 (survival motor neuron 1) but retention of SMN2. The number of copies of SMN2 modifies disease severity in SMA patients as well as in mouse models, making SMN2 a target for therapeutics development. Sodium butyrate (BA) and its analog (4PBA) have been shown to increase SMN2 expression in SMA cultured cells. In this study, we examined the effects of BA, 4PBA as well as two BA prodrugs-glyceryl tributyrate (BA3G) and VX563-on the phenotype of SMNΔ7 SMA mice. Treatment with 4PBA, BA3G and VX563 but not BA beginning at PND04 significantly improved the lifespan and delayed disease end stage, with administration of VX563 also improving the growth rate of these mice. 4PBA and VX563 improved the motor phenotype of SMNΔ7 SMA mice and prevented spinal motor neuron loss. Interestingly, neither 4PBA nor VX563 had an effect on SMN expression in the spinal cords of treated SMNΔ7 SMA mice; however, they inhibited histone deacetylase (HDAC) activity and restored the normal phosphorylation states of Akt and glycogen synthase kinase 3β, both of which are altered by SMN deficiency in vivo. These observations show that BA-based compounds with favorable pharmacokinetics ameliorate SMA pathology possibly by modulating HDAC and Akt signaling.
Collapse
Affiliation(s)
- Matthew E R Butchbach
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Center for Applied Clinical Genomics, Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, DE, USA; Center for Pediatric Research, Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, DE, USA; Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA, USA; Department of Biological Sciences, University of Delaware, Newark, DE, USA.
| | - Casey J Lumpkin
- Center for Applied Clinical Genomics, Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, DE, USA; Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - Ashlee W Harris
- Center for Applied Clinical Genomics, Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Luciano Saieva
- Center for Motor Neuron Biology and Disease, Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Jonathan D Edwards
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Eileen Workman
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Louise R Simard
- Department of Biochemistry and Medical Genetics, University of Manitoba Faculty of Health Sciences, Winnipeg, Manitoba, Canada
| | - Livio Pellizzoni
- Center for Motor Neuron Biology and Disease, Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Arthur H M Burghes
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
333
|
Lin TL, Chen TH, Hsu YY, Cheng YH, Juang BT, Jong YJ. Selective Neuromuscular Denervation in Taiwanese Severe SMA Mouse Can Be Reversed by Morpholino Antisense Oligonucleotides. PLoS One 2016; 11:e0154723. [PMID: 27124114 PMCID: PMC4849667 DOI: 10.1371/journal.pone.0154723] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 04/18/2016] [Indexed: 11/27/2022] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive motor neuron disease caused by deficiency of the survival of motor neuron (SMN) protein, which leads to synaptic defects and spinal motor neuron death. Neuromuscular junction (NMJ) abnormalities have been found to be involved in SMA pathogenesis in the SMNΔ7 SMA mouse model. However, whether similar NMJ pathological findings present in another commonly used mouse model, the Taiwanese SMA mouse, has not been fully investigated. To examine the NMJs of the Taiwanese severe SMA mouse model (Smn-/-; SMN2tg/0), which is characterized by severe phenotype and death before postnatal day (P) 9, we investigated 25 axial and appendicular muscles from P1 to P9. We labelled the muscles with anti-neurofilament and anti-synaptophysin antibodies for nerve terminals and α-bungarotoxin for acetylcholine receptors (AChRs). We found that severe NMJ denervation (<50% fully innervated endplates) selectively occurred in the flexor digitorum brevis 2 and 3 (FDB-2/3) muscles from P5, and an increased percentage of fully denervated endplates correlated with SMA progression. Furthermore, synaptophysin signals were absent at the endplate compared to control littermate mice, suggesting that vesicle transport might only be affected at the end stage. Subsequently, we treated the Taiwanese severe SMA mice with morpholino (MO) antisense oligonucleotides (80 μg/g) via subcutaneous injection at P0. We found that MO significantly reversed the NMJ denervation in FDB-2/3 muscles and extended the survival of Taiwanese severe SMA mice. We conclude that early NMJ denervation in the FDB-2/3 muscles of Taiwanese severe SMA mice can be reversed by MO treatment. The FDB-2/3 muscles of Taiwanese severe SMA mice provide a very sensitive platform for assessing the effectiveness of drug treatments in SMA preclinical studies.
Collapse
Affiliation(s)
- Te-Lin Lin
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tai-Heng Chen
- Division of Pediatric Emergency, Department of Emergency, Kaohsiung Medical University and Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Ya-Yun Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Hua Cheng
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Bi-Tzen Juang
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Yuh-Jyh Jong
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Institute of Molecular Medicine and Bioengineering, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
- Departments of Pediatrics and Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
334
|
Therapeutic activity of modified U1 core spliceosomal particles. Nat Commun 2016; 7:11168. [PMID: 27041075 PMCID: PMC4822034 DOI: 10.1038/ncomms11168] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 02/25/2016] [Indexed: 12/15/2022] Open
Abstract
Modified U1 snRNAs bound to intronic sequences downstream of the 5′ splice site correct exon skipping caused by different types of mutations. Here we evaluate the therapeutic activity and structural requirements of these exon-specific U1 snRNA (ExSpeU1) particles. In a severe spinal muscular atrophy, mouse model, ExSpeU1, introduced by germline transgenesis, increases SMN2 exon 7 inclusion, SMN protein production and extends life span. In vitro, RNA mutant analysis and silencing experiments show that while U1A protein is dispensable, the 70K and stem loop IV elements mediate most of the splicing rescue activity through improvement of exon and intron definition. Our findings indicate that precise engineering of the U1 core spliceosomal RNA particle has therapeutic potential in pathologies associated with exon-skipping mutations. Modification of the spliceosome is being tested as a potential therapy for exon-skipping diseases, such as spinal muscular atrophy (SMA). Here the authors show that 70K and stem loop IV structural elements of a modified U1 particle are essential for splicing enhancement and effective treatment of SMA mice.
Collapse
|
335
|
Liu L, Chen L, Zhang YH, Wei L, Cheng S, Kong X, Zheng M, Huang T, Cai YD. Analysis and prediction of drug-drug interaction by minimum redundancy maximum relevance and incremental feature selection. J Biomol Struct Dyn 2016; 35:312-329. [PMID: 26750516 DOI: 10.1080/07391102.2016.1138142] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Drug-drug interaction (DDI) defines a situation in which one drug affects the activity of another when both are administered together. DDI is a common cause of adverse drug reactions and sometimes also leads to improved therapeutic effects. Therefore, it is of great interest to discover novel DDIs according to their molecular properties and mechanisms in a robust and rigorous way. This paper attempts to predict effective DDIs using the following properties: (1) chemical interaction between drugs; (2) protein interactions between the targets of drugs; and (3) target enrichment of KEGG pathways. The data consisted of 7323 pairs of DDIs collected from the DrugBank and 36,615 pairs of drugs constructed by randomly combining two drugs. Each drug pair was represented by 465 features derived from the aforementioned three categories of properties. The random forest algorithm was adopted to train the prediction model. Some feature selection techniques, including minimum redundancy maximum relevance and incremental feature selection, were used to extract key features as the optimal input for the prediction model. The extracted key features may help to gain insights into the mechanisms of DDIs and provide some guidelines for the relevant clinical medication developments, and the prediction model can give new clues for identification of novel DDIs.
Collapse
Affiliation(s)
- Lili Liu
- a Intelligence Research Department, Information Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , P. R. China
| | - Lei Chen
- b College of Information Engineering, Shanghai Maritime University , Shanghai 201306 , P. R. China
| | - Yu-Hang Zhang
- c Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences , Shanghai 200031 , P. R. China
| | - Lai Wei
- b College of Information Engineering, Shanghai Maritime University , Shanghai 201306 , P. R. China
| | - Shiwen Cheng
- b College of Information Engineering, Shanghai Maritime University , Shanghai 201306 , P. R. China
| | - Xiangyin Kong
- c Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences , Shanghai 200031 , P. R. China
| | - Mingyue Zheng
- d State Key Laboratory of Drug Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , P. R. China
| | - Tao Huang
- c Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences , Shanghai 200031 , P. R. China
| | - Yu-Dong Cai
- e School of Life Sciences, Shanghai University , Shanghai 200444 , P. R. China
| |
Collapse
|
336
|
Ahmad S, Bhatia K, Kannan A, Gangwani L. Molecular Mechanisms of Neurodegeneration in Spinal Muscular Atrophy. J Exp Neurosci 2016; 10:39-49. [PMID: 27042141 PMCID: PMC4807884 DOI: 10.4137/jen.s33122] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 02/22/2016] [Accepted: 02/25/2016] [Indexed: 02/07/2023] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive motor neuron disease with a high incidence and is the most common genetic cause of infant mortality. SMA is primarily characterized by degeneration of the spinal motor neurons that leads to skeletal muscle atrophy followed by symmetric limb paralysis, respiratory failure, and death. In humans, mutation of the Survival Motor Neuron 1 (SMN1) gene shifts the load of expression of SMN protein to the SMN2 gene that produces low levels of full-length SMN protein because of alternative splicing, which are sufficient for embryonic development and survival but result in SMA. The molecular mechanisms of the (a) regulation of SMN gene expression and (b) degeneration of motor neurons caused by low levels of SMN are unclear. However, some progress has been made in recent years that have provided new insights into understanding of the cellular and molecular basis of SMA pathogenesis. In this review, we have briefly summarized recent advances toward understanding of the molecular mechanisms of regulation of SMN levels and signaling mechanisms that mediate neurodegeneration in SMA.
Collapse
Affiliation(s)
- Saif Ahmad
- Center of Emphasis in Neurosciences, Texas Tech University Health Sciences Center, El Paso, Texas, USA.; Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Kanchan Bhatia
- Center of Emphasis in Neurosciences, Texas Tech University Health Sciences Center, El Paso, Texas, USA.; Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Annapoorna Kannan
- Center of Emphasis in Neurosciences, Texas Tech University Health Sciences Center, El Paso, Texas, USA.; Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Laxman Gangwani
- Center of Emphasis in Neurosciences, Texas Tech University Health Sciences Center, El Paso, Texas, USA.; Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| |
Collapse
|
337
|
Christinat Y, Pawłowski R, Krek W. jSplice: a high-performance method for accurate prediction of alternative splicing events and its application to large-scale renal cancer transcriptome data. Bioinformatics 2016; 32:2111-9. [PMID: 27153587 DOI: 10.1093/bioinformatics/btw145] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 03/11/2016] [Indexed: 01/01/2023] Open
Abstract
MOTIVATION Alternative splicing represents a prime mechanism of post-transcriptional gene regulation whose misregulation is associated with a broad range of human diseases. Despite the vast availability of transcriptome data from different cell types and diseases, bioinformatics-based surveys of alternative splicing patterns remain a major challenge due to limited availability of analytical tools that combine high accuracy and rapidity. RESULTS We describe here a novel junction-centric method, jSplice, that enables de novo extraction of alternative splicing events from RNA-sequencing data with high accuracy, reliability and speed. Application to clear cell renal carcinoma (ccRCC) cell lines and 65 ccRCC patients revealed experimentally validatable alternative splicing changes and signatures able to prognosticate ccRCC outcome. In the aggregate, our results propose jSplice as a key analytic tool for the derivation of cell context-dependent alternative splicing patterns from large-scale RNA-sequencing datasets. AVAILABILITY AND IMPLEMENTATION jSplice is a standalone Python application freely available at http://www.mhs.biol.ethz.ch/research/krek/jsplice CONTACT wilhelm.krek@biol.ethz.ch SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Yann Christinat
- Institute of Molecular Health Sciences, ETH Zurich, Zurich 8093, Switzerland
| | - Rafał Pawłowski
- Institute of Molecular Health Sciences, ETH Zurich, Zurich 8093, Switzerland
| | - Wilhelm Krek
- Institute of Molecular Health Sciences, ETH Zurich, Zurich 8093, Switzerland
| |
Collapse
|
338
|
Mavrou A, Oltean S. SRPK1 inhibition in prostate cancer: A novel anti-angiogenic treatment through modulation of VEGF alternative splicing. Pharmacol Res 2016; 107:276-281. [PMID: 26995304 PMCID: PMC4876767 DOI: 10.1016/j.phrs.2016.03.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 03/11/2016] [Indexed: 02/02/2023]
Abstract
Prostate cancer remains one of the leading causes of cancer death in men around the world, regardless of intense research and development of novel therapies in the last 10 years. One of the new avenues that has been tested - inhibition of angiogenesis - has been disappointing so far in clinical studies in spite of strong evidence that determinants of angiogenesis (e.g. vascular endothelial growth factor) are strongly associated with disease progression. One of the reasons for these outcomes may be our poor understanding of the biology of angiogenesis in prostate cancer (and probably other cancers as well) resulting in inhibition of both detrimental and favourable molecules. We discuss here novel targeted and more specific approaches to inhibit angiogenesis in prostate cancer as well as a completely new therapeutic modality to do this - modulation of alternative splicing - that may be applicable to other molecules/biological processes as well.
Collapse
Affiliation(s)
- Athina Mavrou
- School of Physiology, Pharmacology and Neurosciences, UK
| | - Sebastian Oltean
- School of Physiology, Pharmacology and Neurosciences, UK; School of Clinical Sciences/Bristol Renal, University of Bristol, UK.
| |
Collapse
|
339
|
Zaworski P, von Herrmann KM, Taylor S, Sunshine SS, McCarthy K, Risher N, Newcomb T, Weetall M, Prior TW, Swoboda KJ, Chen KS, Paushkin S. SMN Protein Can Be Reliably Measured in Whole Blood with an Electrochemiluminescence (ECL) Immunoassay: Implications for Clinical Trials. PLoS One 2016; 11:e0150640. [PMID: 26953792 PMCID: PMC4783032 DOI: 10.1371/journal.pone.0150640] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 02/17/2016] [Indexed: 11/18/2022] Open
Abstract
Spinal muscular atrophy (SMA) is caused by defects in the survival motor neuron 1 (SMN1) gene that encodes survival motor neuron (SMN) protein. The majority of therapeutic approaches currently in clinical development for SMA aim to increase SMN protein expression and there is a need for sensitive methods able to quantify increases in SMN protein levels in accessible tissues. We have developed a sensitive electrochemiluminescence (ECL)-based immunoassay for measuring SMN protein in whole blood with a minimum volume requirement of 5μL. The SMN-ECL immunoassay enables accurate measurement of SMN in whole blood and other tissues. Using the assay, we measured SMN protein in whole blood from SMA patients and healthy controls and found that SMN protein levels were associated with SMN2 copy number and were greater in SMA patients with 4 copies, relative to those with 2 and 3 copies. SMN protein levels did not vary significantly in healthy individuals over a four-week period and were not affected by circadian rhythms. Almost half of the SMN protein was found in platelets. We show that SMN protein levels in C/C-allele mice, which model a mild form of SMA, were high in neonatal stage, decreased in the first few weeks after birth, and then remained stable throughout the adult stage. Importantly, SMN protein levels in the CNS correlated with SMN levels measured in whole blood of the C/C-allele mice. These findings have implications for the measurement of SMN protein induction in whole blood in response to SMN-upregulating therapy.
Collapse
Affiliation(s)
| | | | - Shannon Taylor
- PharmOptima, Portage, Michigan, United States of America
| | - Sara S. Sunshine
- Spinal Muscular Atrophy Foundation, New York, New York, United States of America
| | - Kathleen McCarthy
- Spinal Muscular Atrophy Foundation, New York, New York, United States of America
| | - Nicole Risher
- PTC Therapeutics, South Plainfield, New Jersey, United States of America
| | - Tara Newcomb
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Marla Weetall
- PTC Therapeutics, South Plainfield, New Jersey, United States of America
| | - Thomas W. Prior
- Department of Molecular Pathology, Wexner Medical Center, Ohio State University, Columbus, Ohio, United States of America
| | - Kathryn J. Swoboda
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Karen S. Chen
- Spinal Muscular Atrophy Foundation, New York, New York, United States of America
| | - Sergey Paushkin
- Spinal Muscular Atrophy Foundation, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
340
|
Zhao X, Feng Z, Ling KKY, Mollin A, Sheedy J, Yeh S, Petruska J, Narasimhan J, Dakka A, Welch EM, Karp G, Chen KS, Metzger F, Ratni H, Lotti F, Tisdale S, Naryshkin NA, Pellizzoni L, Paushkin S, Ko CP, Weetall M. Pharmacokinetics, pharmacodynamics, and efficacy of a small-molecule SMN2 splicing modifier in mouse models of spinal muscular atrophy. Hum Mol Genet 2016; 25:1885-1899. [PMID: 26931466 PMCID: PMC5062580 DOI: 10.1093/hmg/ddw062] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/22/2016] [Indexed: 12/26/2022] Open
Abstract
Spinal muscular atrophy (SMA) is caused by the loss or mutation of both copies of the survival motor neuron 1 (SMN1) gene. The related SMN2 gene is retained, but due to alternative splicing of exon 7, produces insufficient levels of the SMN protein. Here, we systematically characterize the pharmacokinetic and pharmacodynamics properties of the SMN splicing modifier SMN-C1. SMN-C1 is a low-molecular weight compound that promotes the inclusion of exon 7 and increases production of SMN protein in human cells and in two transgenic mouse models of SMA. Furthermore, increases in SMN protein levels in peripheral blood mononuclear cells and skin correlate with those in the central nervous system (CNS), indicating that a change of these levels in blood or skin can be used as a non-invasive surrogate to monitor increases of SMN protein levels in the CNS. Consistent with restored SMN function, SMN-C1 treatment increases the levels of spliceosomal and U7 small-nuclear RNAs and corrects RNA processing defects induced by SMN deficiency in the spinal cord of SMNΔ7 SMA mice. A 100% or greater increase in SMN protein in the CNS of SMNΔ7 SMA mice robustly improves the phenotype. Importantly, a ∼50% increase in SMN leads to long-term survival, but the SMA phenotype is only partially corrected, indicating that certain SMA disease manifestations may respond to treatment at lower doses. Overall, we provide important insights for the translation of pre-clinical data to the clinic and further therapeutic development of this series of molecules for SMA treatment.
Collapse
Affiliation(s)
- Xin Zhao
- PTC Therapeutics, Inc., South Plainfield, NJ 07080, USA
| | - Zhihua Feng
- Department of Biological Sciences, Section of Neurobiology, University of Southern California, Los Angeles, CA 90089, USA
| | - Karen K Y Ling
- Department of Biological Sciences, Section of Neurobiology, University of Southern California, Los Angeles, CA 90089, USA
| | - Anna Mollin
- PTC Therapeutics, Inc., South Plainfield, NJ 07080, USA
| | | | - Shirley Yeh
- PTC Therapeutics, Inc., South Plainfield, NJ 07080, USA
| | | | | | - Amal Dakka
- PTC Therapeutics, Inc., South Plainfield, NJ 07080, USA
| | - Ellen M Welch
- PTC Therapeutics, Inc., South Plainfield, NJ 07080, USA
| | - Gary Karp
- PTC Therapeutics, Inc., South Plainfield, NJ 07080, USA
| | - Karen S Chen
- SMA Foundation, 888 Seventh Avenue, Suite 400, New York, NY 10019, USA
| | - Friedrich Metzger
- F. Hoffmann-La Roche, Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Hasane Ratni
- F. Hoffmann-La Roche, Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Francesco Lotti
- Department of Pathology and Cell Biology, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA and
| | - Sarah Tisdale
- Department of Pathology and Cell Biology, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA and
| | | | - Livio Pellizzoni
- Department of Pathology and Cell Biology, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA and
| | - Sergey Paushkin
- SMA Foundation, 888 Seventh Avenue, Suite 400, New York, NY 10019, USA
| | - Chien-Ping Ko
- Department of Biological Sciences, Section of Neurobiology, University of Southern California, Los Angeles, CA 90089, USA,
| | - Marla Weetall
- PTC Therapeutics, Inc., South Plainfield, NJ 07080, USA,
| |
Collapse
|
341
|
Iijima T, Hidaka C, Iijima Y. Spatio-temporal regulations and functions of neuronal alternative RNA splicing in developing and adult brains. Neurosci Res 2016; 109:1-8. [PMID: 26853282 DOI: 10.1016/j.neures.2016.01.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 01/25/2016] [Accepted: 01/25/2016] [Indexed: 10/25/2022]
Abstract
Alternative pre-mRNA splicing is a fundamental mechanism that generates molecular diversity from a single gene. In the central nervous system (CNS), key neural developmental steps are thought to be controlled by alternative splicing decisions, including the molecular diversity underlying synaptic wiring, plasticity, and remodeling. Significant progress has been made in understanding the molecular mechanisms and functions of alternative pre-mRNA splicing in neurons through studies in invertebrate systems; however, recent studies have begun to uncover the potential role of neuronal alternative splicing in the mammalian CNS. This article provides an overview of recent findings regarding the regulation and function of neuronal alternative splicing. In particular, we focus on the spatio-temporal regulation of neurexin, a synaptic adhesion molecule, by neuronal cell type-specific factors and neuronal activity, which are thought to be especially important for characterizing neural development and function within the mammalian CNS. Notably, there is increasing evidence that implicates the dysregulation of neuronal splicing events in several neurological disorders. Therefore, understanding the detailed mechanisms of neuronal alternative splicing in the mammalian CNS may provide plausible treatment strategies for these diseases.
Collapse
Affiliation(s)
- Takatoshi Iijima
- Tokai University Institute of Innovative Science and Technology, 143 Shimokasuya, Isehara City, Kanagawa 259-1193, Japan; Tokai University Institute of Innovative Science and Technology, 4-1-1 Kitakaname, Hiratsuka City, Kanagawa 259-1292, Japan; School of Medicine, Tokai University, 143 Shimokasuya, Isehara City, Kanagawa 259-1193, Japan.
| | - Chiharu Hidaka
- Tokai University Institute of Innovative Science and Technology, 143 Shimokasuya, Isehara City, Kanagawa 259-1193, Japan; Tokai University Institute of Innovative Science and Technology, 4-1-1 Kitakaname, Hiratsuka City, Kanagawa 259-1292, Japan; School of Medicine, Tokai University, 143 Shimokasuya, Isehara City, Kanagawa 259-1193, Japan
| | - Yoko Iijima
- Tokai University Institute of Innovative Science and Technology, 143 Shimokasuya, Isehara City, Kanagawa 259-1193, Japan; Tokai University Institute of Innovative Science and Technology, 4-1-1 Kitakaname, Hiratsuka City, Kanagawa 259-1292, Japan; School of Medicine, Tokai University, 143 Shimokasuya, Isehara City, Kanagawa 259-1193, Japan
| |
Collapse
|
342
|
Frank AT. Can Holo NMR Chemical Shifts be Directly Used to Resolve RNA–Ligand Poses? J Chem Inf Model 2016; 56:368-76. [DOI: 10.1021/acs.jcim.5b00593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Aaron T. Frank
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
343
|
Abstract
Alternative splicing (AS) has emerged in the postgenomic era as one of the main drivers of proteome diversity, with ≥94% of multiexon genes alternatively spliced in humans. AS is therefore one of the main control mechanisms for cell phenotype, and is a process deregulated in disease. Numerous reports describe pathogenic mutations in splice factors, splice sites, or regulatory sequences. Additionally, compared with the physiologic state, disease often associates with an abnormal proportion of splice isoforms (or novel isoforms), without an apparent driver mutation. It is therefore essential to study how AS is regulated in physiology, how it contributes to pathogenesis, and whether we can manipulate faulty splicing for therapeutic advantage. Although the disease most commonly linked to deregulation of AS in several genes is cancer, many reports detail pathogenic splice variants in diseases ranging from neuromuscular disorders to diabetes or cardiomyopathies. A plethora of splice variants have been implicated in CKDs as well. In this review, we describe examples of these CKD-associated splice variants and ideas on how to manipulate them for therapeutic benefit.
Collapse
Affiliation(s)
- Megan Stevens
- School of Physiology and Pharmacology, Faculty of Biomedical Sciences, and Academic Renal Unit, School of Clinical Sciences, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Sebastian Oltean
- School of Physiology and Pharmacology, Faculty of Biomedical Sciences, and Academic Renal Unit, School of Clinical Sciences, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
344
|
Feng Z, Ling KKY, Zhao X, Zhou C, Karp G, Welch EM, Naryshkin N, Ratni H, Chen KS, Metzger F, Paushkin S, Weetall M, Ko CP. Pharmacologically induced mouse model of adult spinal muscular atrophy to evaluate effectiveness of therapeutics after disease onset. Hum Mol Genet 2016; 25:964-75. [PMID: 26758873 DOI: 10.1093/hmg/ddv629] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 12/29/2015] [Indexed: 12/21/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a genetic disease characterized by atrophy of muscle and loss of spinal motor neurons. SMA is caused by deletion or mutation of the survival motor neuron 1 (SMN1) gene, and the nearly identical SMN2 gene fails to generate adequate levels of functional SMN protein due to a splicing defect. Currently, several therapeutics targeted to increase SMN protein are in clinical trials. An outstanding issue in the field is whether initiating treatment in symptomatic older patients would confer a therapeutic benefit, an important consideration as the majority of patients with milder forms of SMA are diagnosed at an older age. An SMA mouse model that recapitulates the disease phenotype observed in adolescent and adult SMA patients is needed to address this important question. We demonstrate here that Δ7 mice, a model of severe SMA, treated with a suboptimal dose of an SMN2 splicing modifier show increased SMN protein, survive into adulthood and display SMA disease-relevant pathologies. Increasing the dose of the splicing modifier after the disease symptoms are apparent further mitigates SMA histopathological features in suboptimally dosed adult Δ7 mice. In addition, inhibiting myostatin using intramuscular injection of AAV1-follistatin ameliorates muscle atrophy in suboptimally dosed Δ7 mice. Taken together, we have developed a new murine model of symptomatic SMA in adolescents and adult mice that is induced pharmacologically from a more severe model and demonstrated efficacy of both SMN2 splicing modifiers and a myostatin inhibitor in mice at later disease stages.
Collapse
Affiliation(s)
- Zhihua Feng
- Section of Neurobiology, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089-2520, USA
| | - Karen K Y Ling
- Section of Neurobiology, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089-2520, USA
| | - Xin Zhao
- PTC Therapeutics, Inc., South Plainfield, NJ 07080, USA
| | - Chunyi Zhou
- Section of Neurobiology, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089-2520, USA
| | - Gary Karp
- PTC Therapeutics, Inc., South Plainfield, NJ 07080, USA
| | - Ellen M Welch
- PTC Therapeutics, Inc., South Plainfield, NJ 07080, USA
| | | | - Hasane Ratni
- F. Hoffmann-La Roche Ltd, Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland and
| | - Karen S Chen
- SMA Foundation, 888 Seventh Avenue, Suite 400, New York, NY 10019, USA
| | - Friedrich Metzger
- F. Hoffmann-La Roche Ltd, Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland and
| | - Sergey Paushkin
- SMA Foundation, 888 Seventh Avenue, Suite 400, New York, NY 10019, USA
| | - Marla Weetall
- PTC Therapeutics, Inc., South Plainfield, NJ 07080, USA
| | - Chien-Ping Ko
- Section of Neurobiology, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089-2520, USA,
| |
Collapse
|
345
|
Introduction. Transl Neurosci 2016. [DOI: 10.1007/978-1-4899-7654-3_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
346
|
Arnold W, McGovern VL, Sanchez B, Li J, Corlett KM, Kolb SJ, Rutkove SB, Burghes AH. The neuromuscular impact of symptomatic SMN restoration in a mouse model of spinal muscular atrophy. Neurobiol Dis 2015; 87:116-23. [PMID: 26733414 DOI: 10.1016/j.nbd.2015.12.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 12/20/2015] [Accepted: 12/25/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Significant advances in the development of SMN-restoring therapeutics have occurred since 2010 when very effective biological treatments were reported in mouse models of spinal muscular atrophy. As these treatments are applied in human clinical trials, there is pressing need to define quantitative assessments of disease progression, treatment stratification, and therapeutic efficacy. The electrophysiological measures Compound Muscle Action Potential and Motor Unit Number Estimation are reliable measures of nerve function. In both the SMN∆7 mouse and a pig model of spinal muscular atrophy, early SMN restoration results in preservation of electrophysiological measures. Currently, clinical trials are underway in patients at post-symptomatic stages of disease progression. In this study, we present results from both early and delayed SMN restoration using clinically-relevant measures including electrical impedance myography, compound muscle action potential, and motor unit number estimation to quantify the efficacy and time-sensitivity of SMN-restoring therapy. METHODS SMA∆7 mice were treated via intracerebroventricular injection with antisense oligonucleotides targeting ISS-N1 to increase SMN protein from the SMN2 gene on postnatal day 2, 4, or 6 and compared with sham-treated spinal muscular atrophy and control mice. Compound muscle action potential and motor unit number estimation of the triceps surae muscles were performed at day 12, 21, and 30 by a single evaluator blinded to genotype and treatment. Similarly, electrical impedance myography was measured on the biceps femoris muscle at 12days for comparison. RESULTS Electrophysiological measures and electrical impedance myography detected significant differences at 12days between control and late-treated (4 or 6days) and sham-treated spinal muscular atrophy mice, but not in mice treated at 2days (p<0.01). EIM findings paralleled and correlated with compound muscle action potential and motor unit number estimation (r=0.61 and r=0.50, respectively, p<0.01). Longitudinal measures at 21 and 30days show that symptomatic therapy results in reduced motor unit number estimation associated with delayed normalization of compound muscle action potential. CONCLUSIONS The incomplete effect of symptomatic treatment is accurately identified by both electrophysiological measures and electrical impedance myography. There is strong correlation between these measures and with weight and righting reflex. This study predicts that measures of compound muscle action potential, motor unit number estimation, and electrical impedance myography are promising biomarkers of treatment stratification and effect for future spinal muscular atrophy trials. The ease of application and simplicity of electrical impedance myography compared with standard electrophysiological measures may be particularly valuable in future pediatric clinical trials.
Collapse
Affiliation(s)
- W Arnold
- Department of Neurology, The Ohio State University Wexner Medical Center, 395 W. 12th Ave, Columbus, OH 43210, United States; Department of Physical Medicine and Rehabilitation, The Ohio State University Wexner Medical Center, 480 Medical Center Drive, Columbus, OH 43210, United States; Department of Neuroscience, The Ohio State University Wexner Medical Center, 480 Medical Center Drive, Columbus, OH 43210, United States
| | - Vicki L McGovern
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, 363 Hamilton Hall, 1645 Neil Ave, Columbus, OH 43210, United States
| | - Benjamin Sanchez
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States
| | - Jia Li
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States
| | - Kaitlyn M Corlett
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, 363 Hamilton Hall, 1645 Neil Ave, Columbus, OH 43210, United States
| | - Stephen J Kolb
- Department of Neurology, The Ohio State University Wexner Medical Center, 395 W. 12th Ave, Columbus, OH 43210, United States; Department of Neuroscience, The Ohio State University Wexner Medical Center, 480 Medical Center Drive, Columbus, OH 43210, United States; Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, 363 Hamilton Hall, 1645 Neil Ave, Columbus, OH 43210, United States
| | - Seward B Rutkove
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States
| | - Arthur H Burghes
- Department of Neurology, The Ohio State University Wexner Medical Center, 395 W. 12th Ave, Columbus, OH 43210, United States; Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, 363 Hamilton Hall, 1645 Neil Ave, Columbus, OH 43210, United States
| |
Collapse
|
347
|
Ohuchi K, Funato M, Kato Z, Seki J, Kawase C, Tamai Y, Ono Y, Nagahara Y, Noda Y, Kameyama T, Ando S, Tsuruma K, Shimazawa M, Hara H, Kaneko H. Established Stem Cell Model of Spinal Muscular Atrophy Is Applicable in the Evaluation of the Efficacy of Thyrotropin-Releasing Hormone Analog. Stem Cells Transl Med 2015; 5:152-63. [PMID: 26683872 DOI: 10.5966/sctm.2015-0059] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 10/07/2015] [Indexed: 11/16/2022] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive neuromuscular disorder characterized by the degeneration of spinal motor neurons. This disease is mainly caused by mutation or deletion of the survival motor neuron 1 (SMN1) gene. Currently, no effective treatment is available, and only symptomatic treatment can be provided. Our purpose in the present study was to establish a human SMA-derived induced pluripotent stem cell (SMA-iPSC) disease model and assay a therapeutic drug in preparation for the development of a novel treatment of SMA. We generated iPSCs from the skin fibroblasts of a patient with SMA and confirmed that they were pluripotent and undifferentiated. The neural differentiation of SMA-iPSCs shortened the dendrite and axon length and increased the apoptosis of the spinal motor neurons. In addition, we found activated astrocytes in differentiated SMA-iPSCs. Using this model, we confirmed that treatment with the thyrotropin-releasing hormone (TRH) analog, 5-oxo-l-prolyl-l-histidyl-l-prolinamide, which had marginal effects in clinical trials, increases the SMN protein level. This increase was mediated through the transcriptional activation of the SMN2 gene and inhibition of glycogen synthase kinase-3β activity. Finally, the TRH analog treatment resulted in dendrite and axon development of spinal motor neurons in differentiated SMA-iPSCs. These results suggest that this human in vitro disease model stimulates SMA pathology and reveal the potential efficacy of TRH analog treatment for SMA. Therefore, we can screen novel therapeutic drugs such as TRH for SMA easily and effectively using the human SMA-iPSC model. Significance: Platelet-derived growth factor (PDGF) has recently been reported to produce the greatest increase in survival motor neuron protein levels by inhibiting glycogen synthase kinase (GSK)-3β; however, motor neurons lack PDGF receptors. A human in vitro spinal muscular atrophy-derived induced pluripotent stem cell model was established, which showed that the thyrotropin releasing hormone (TRH) analog promoted transcriptional activation of the SMN2 gene and inhibition of GSK-3β activity, resulting in the increase and stabilization of the SMN protein and axon elongation of spinal motor neurons. These results reveal the potential efficacy of TRH analog treatment for SMA.
Collapse
Affiliation(s)
- Kazuki Ohuchi
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Michinori Funato
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Zenichiro Kato
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, Japan
| | - Junko Seki
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Chizuru Kawase
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Yuya Tamai
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Yoko Ono
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Yuki Nagahara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Yasuhiro Noda
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Tsubasa Kameyama
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Shiori Ando
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Kazuhiro Tsuruma
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Hideo Kaneko
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| |
Collapse
|
348
|
Abstract
The human transcriptome is composed of a vast RNA population that undergoes further diversification by splicing. Detecting specific splice sites in this large sequence pool is the responsibility of the major and minor spliceosomes in collaboration with numerous splicing factors. This complexity makes splicing susceptible to sequence polymorphisms and deleterious mutations. Indeed, RNA mis-splicing underlies a growing number of human diseases with substantial societal consequences. Here, we provide an overview of RNA splicing mechanisms followed by a discussion of disease-associated errors, with an emphasis on recently described mutations that have provided new insights into splicing regulation. We also discuss emerging strategies for splicing-modulating therapy.
Collapse
Affiliation(s)
- Marina M Scotti
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, Florida 32610-3610 USA
| | - Maurice S Swanson
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, Florida 32610-3610 USA
| |
Collapse
|
349
|
Zhou H, Meng J, Marrosu E, Janghra N, Morgan J, Muntoni F. Repeated low doses of morpholino antisense oligomer: an intermediate mouse model of spinal muscular atrophy to explore the window of therapeutic response. Hum Mol Genet 2015; 24:6265-77. [PMID: 26264577 PMCID: PMC4614699 DOI: 10.1093/hmg/ddv329] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 07/20/2015] [Accepted: 08/06/2015] [Indexed: 11/14/2022] Open
Abstract
The human SMN2 transgenic mice are well-established models of spinal muscular atrophy (SMA). While the severe type I mouse model has a rapidly progressive condition mimicking type I SMA in humans, the mild type III mice do not faithfully recapitulate chronic SMA variants affecting children. A SMA mouse model that clinically mimics the features of type II and III SMA in human is therefore needed. In this study, we generated intermediately affected SMA mice by delivering low-dose morpholino oligomer (PMO25) into the existing severe SMA mice. We show that a single low-dose administration of PMO25 moderately extended the survival of severe type I SMA mice. The neuromuscular pathology is also modestly but significantly improved in these mice. A second administration of PMO25 at postnatal day 5 (PND5) demonstrated an additive effect on survival. Additional systemic administration of low-dose PMO25 at 2-week intervals suppressed the occurrence of distal necrosis beyond postnatal day 100, and induced more complete phenotypic rescue than a single bolus high-dose injection at PND0. Our study demonstrates that survival of motor neuron (SMN) is required early at a critical threshold to prevent symptoms and suggests that subsequent systemic administration of low-dose PMO25 in SMA mice can provide therapeutic benefit and phenotypic rescue, presumably via peripheral SMN restoration. Our work also provides additional insight into the time window of response to administration of antisense oligonucleotides to SMA mice with an intermediate phenotype. This information is crucial at a time when a number of therapeutic interventions are in clinical trials in SMA patients.
Collapse
Affiliation(s)
- Haiyan Zhou
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Jinhong Meng
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Elena Marrosu
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Narinder Janghra
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Jennifer Morgan
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
350
|
Daguenet E, Dujardin G, Valcárcel J. The pathogenicity of splicing defects: mechanistic insights into pre-mRNA processing inform novel therapeutic approaches. EMBO Rep 2015; 16:1640-55. [PMID: 26566663 DOI: 10.15252/embr.201541116] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/12/2015] [Indexed: 12/22/2022] Open
Abstract
Removal of introns from pre-mRNA precursors (pre-mRNA splicing) is a necessary step for the expression of most genes in multicellular organisms, and alternative patterns of intron removal diversify and regulate the output of genomic information. Mutation or natural variation in pre-mRNA sequences, as well as in spliceosomal components and regulatory factors, has been implicated in the etiology and progression of numerous pathologies. These range from monogenic to multifactorial genetic diseases, including metabolic syndromes, muscular dystrophies, neurodegenerative and cardiovascular diseases, and cancer. Understanding the molecular mechanisms associated with splicing-related pathologies can provide key insights into the normal function and physiological context of the complex splicing machinery and establish sound basis for novel therapeutic approaches.
Collapse
Affiliation(s)
- Elisabeth Daguenet
- Centre de Regulació Genòmica (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain Universitat Pompeu-Fabra, Barcelona, Spain
| | - Gwendal Dujardin
- Centre de Regulació Genòmica (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain Universitat Pompeu-Fabra, Barcelona, Spain
| | - Juan Valcárcel
- Centre de Regulació Genòmica (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain Universitat Pompeu-Fabra, Barcelona, Spain Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|