LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 7 of total 7

Search options

  1. Article ; Online: Development of an oral treatment that rescues gait ataxia and retinal degeneration in a phenotypic mouse model of familial dysautonomia.

    Morini, Elisabetta / Chekuri, Anil / Logan, Emily M / Bolduc, Jessica M / Kirchner, Emily G / Salani, Monica / Krauson, Aram J / Narasimhan, Jana / Gabbeta, Vijayalakshmi / Grover, Shivani / Dakka, Amal / Mollin, Anna / Jung, Stephen P / Zhao, Xin / Zhang, Nanjing / Zhang, Sophie / Arnold, Michael / Woll, Matthew G / Naryshkin, Nikolai A /
    Weetall, Marla / Slaugenhaupt, Susan A

    American journal of human genetics

    2023  Volume 110, Issue 3, Page(s) 531–547

    Abstract: Familial dysautonomia (FD) is a rare neurodegenerative disease caused by a splicing mutation in elongator acetyltransferase complex subunit 1 (ELP1). This mutation leads to the skipping of exon 20 and a tissue-specific reduction of ELP1, mainly in the ... ...

    Abstract Familial dysautonomia (FD) is a rare neurodegenerative disease caused by a splicing mutation in elongator acetyltransferase complex subunit 1 (ELP1). This mutation leads to the skipping of exon 20 and a tissue-specific reduction of ELP1, mainly in the central and peripheral nervous systems. FD is a complex neurological disorder accompanied by severe gait ataxia and retinal degeneration. There is currently no effective treatment to restore ELP1 production in individuals with FD, and the disease is ultimately fatal. After identifying kinetin as a small molecule able to correct the ELP1 splicing defect, we worked on its optimization to generate novel splicing modulator compounds (SMCs) that can be used in individuals with FD. Here, we optimize the potency, efficacy, and bio-distribution of second-generation kinetin derivatives to develop an oral treatment for FD that can efficiently pass the blood-brain barrier and correct the ELP1 splicing defect in the nervous system. We demonstrate that the novel compound PTC258 efficiently restores correct ELP1 splicing in mouse tissues, including brain, and most importantly, prevents the progressive neuronal degeneration that is characteristic of FD. Postnatal oral administration of PTC258 to the phenotypic mouse model TgFD9;Elp1
    MeSH term(s) Mice ; Animals ; Dysautonomia, Familial/genetics ; Kinetin ; Gait Ataxia ; Retinal Degeneration ; Neurodegenerative Diseases ; Administration, Oral
    Chemical Substances Kinetin (P39Y9652YJ)
    Language English
    Publishing date 2023-02-20
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 219384-x
    ISSN 1537-6605 ; 0002-9297
    ISSN (online) 1537-6605
    ISSN 0002-9297
    DOI 10.1016/j.ajhg.2023.01.019
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: A deep learning approach to identify gene targets of a therapeutic for human splicing disorders.

    Gao, Dadi / Morini, Elisabetta / Salani, Monica / Krauson, Aram J / Chekuri, Anil / Sharma, Neeraj / Ragavendran, Ashok / Erdin, Serkan / Logan, Emily M / Li, Wencheng / Dakka, Amal / Narasimhan, Jana / Zhao, Xin / Naryshkin, Nikolai / Trotta, Christopher R / Effenberger, Kerstin A / Woll, Matthew G / Gabbeta, Vijayalakshmi / Karp, Gary /
    Yu, Yong / Johnson, Graham / Paquette, William D / Cutting, Garry R / Talkowski, Michael E / Slaugenhaupt, Susan A

    Nature communications

    2021  Volume 12, Issue 1, Page(s) 3332

    Abstract: Pre-mRNA splicing is a key controller of human gene expression. Disturbances in splicing due to mutation lead to dysregulated protein expression and contribute to a substantial fraction of human disease. Several classes of splicing modulator compounds ( ... ...

    Abstract Pre-mRNA splicing is a key controller of human gene expression. Disturbances in splicing due to mutation lead to dysregulated protein expression and contribute to a substantial fraction of human disease. Several classes of splicing modulator compounds (SMCs) have been recently identified and establish that pre-mRNA splicing represents a target for therapy. We describe herein the identification of BPN-15477, a SMC that restores correct splicing of ELP1 exon 20. Using transcriptome sequencing from treated fibroblast cells and a machine learning approach, we identify BPN-15477 responsive sequence signatures. We then leverage this model to discover 155 human disease genes harboring ClinVar mutations predicted to alter pre-mRNA splicing as targets for BPN-15477. Splicing assays confirm successful correction of splicing defects caused by mutations in CFTR, LIPA, MLH1 and MAPT. Subsequent validations in two disease-relevant cellular models demonstrate that BPN-15477 increases functional protein, confirming the clinical potential of our predictions.
    MeSH term(s) Animals ; Computational Biology ; Cystic Fibrosis Transmembrane Conductance Regulator/genetics ; Deep Learning ; Exons ; Gene Targeting/methods ; HEK293 Cells ; Humans ; Mice ; Mice, Transgenic ; MutL Protein Homolog 1/genetics ; Mutation ; Phenethylamines/administration & dosage ; Pyridazines/administration & dosage ; RNA Splicing ; Sterol Esterase/genetics ; Transcriptome ; tau Proteins/genetics
    Chemical Substances CFTR protein, human ; MAPT protein, human ; MLH1 protein, human ; Phenethylamines ; Pyridazines ; tau Proteins ; Cystic Fibrosis Transmembrane Conductance Regulator (126880-72-6) ; LIPA protein, human (EC 3.1.1.13) ; Sterol Esterase (EC 3.1.1.13) ; MutL Protein Homolog 1 (EC 3.6.1.3)
    Language English
    Publishing date 2021-06-07
    Publishing country England
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 2553671-0
    ISSN 2041-1723 ; 2041-1723
    ISSN (online) 2041-1723
    ISSN 2041-1723
    DOI 10.1038/s41467-021-23663-2
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: Small molecule splicing modifiers with systemic HTT-lowering activity.

    Bhattacharyya, Anuradha / Trotta, Christopher R / Narasimhan, Jana / Wiedinger, Kari J / Li, Wencheng / Effenberger, Kerstin A / Woll, Matthew G / Jani, Minakshi B / Risher, Nicole / Yeh, Shirley / Cheng, Yaofeng / Sydorenko, Nadiya / Moon, Young-Choon / Karp, Gary M / Weetall, Marla / Dakka, Amal / Gabbeta, Vijayalakshmi / Naryshkin, Nikolai A / Graci, Jason D /
    Tripodi, Thomas / Southwell, Amber / Hayden, Michael / Colacino, Joseph M / Peltz, Stuart W

    Nature communications

    2021  Volume 12, Issue 1, Page(s) 7299

    Abstract: Huntington's disease (HD) is a hereditary neurodegenerative disorder caused by expansion of cytosine-adenine-guanine (CAG) trinucleotide repeats in the huntingtin (HTT) gene. Consequently, the mutant protein is ubiquitously expressed and drives ... ...

    Abstract Huntington's disease (HD) is a hereditary neurodegenerative disorder caused by expansion of cytosine-adenine-guanine (CAG) trinucleotide repeats in the huntingtin (HTT) gene. Consequently, the mutant protein is ubiquitously expressed and drives pathogenesis of HD through a toxic gain-of-function mechanism. Animal models of HD have demonstrated that reducing huntingtin (HTT) protein levels alleviates motor and neuropathological abnormalities. Investigational drugs aim to reduce HTT levels by repressing HTT transcription, stability or translation. These drugs require invasive procedures to reach the central nervous system (CNS) and do not achieve broad CNS distribution. Here, we describe the identification of orally bioavailable small molecules with broad distribution throughout the CNS, which lower HTT expression consistently throughout the CNS and periphery through selective modulation of pre-messenger RNA splicing. These compounds act by promoting the inclusion of a pseudoexon containing a premature termination codon (stop-codon psiExon), leading to HTT mRNA degradation and reduction of HTT levels.
    MeSH term(s) Animals ; Central Nervous System/drug effects ; Central Nervous System/metabolism ; Disease Models, Animal ; Humans ; Huntingtin Protein/genetics ; Huntingtin Protein/metabolism ; Huntington Disease/drug therapy ; Huntington Disease/genetics ; Huntington Disease/metabolism ; Mice ; RNA Splicing/drug effects ; RNA Stability/drug effects ; Small Molecule Libraries/administration & dosage ; Trinucleotide Repeat Expansion/drug effects
    Chemical Substances Htt protein, mouse ; Huntingtin Protein ; Small Molecule Libraries
    Language English
    Publishing date 2021-12-15
    Publishing country England
    Document type Journal Article
    ZDB-ID 2553671-0
    ISSN 2041-1723 ; 2041-1723
    ISSN (online) 2041-1723
    ISSN 2041-1723
    DOI 10.1038/s41467-021-27157-z
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Age-dependent SMN expression in disease-relevant tissue and implications for SMA treatment.

    Ramos, Daniel M / d'Ydewalle, Constantin / Gabbeta, Vijayalakshmi / Dakka, Amal / Klein, Stephanie K / Norris, Daniel A / Matson, John / Taylor, Shannon J / Zaworski, Phillip G / Prior, Thomas W / Snyder, Pamela J / Valdivia, David / Hatem, Christine L / Waters, Ian / Gupte, Nikhil / Swoboda, Kathryn J / Rigo, Frank / Bennett, C Frank / Naryshkin, Nikolai /
    Paushkin, Sergey / Crawford, Thomas O / Sumner, Charlotte J

    The Journal of clinical investigation

    2019  Volume 129, Issue 11, Page(s) 4817–4831

    Abstract: BACKGROUNDSpinal muscular atrophy (SMA) is caused by deficient expression of survival motor neuron (SMN) protein. New SMN-enhancing therapeutics are associated with variable clinical benefits. Limited knowledge of baseline and drug-induced SMN levels in ... ...

    Abstract BACKGROUNDSpinal muscular atrophy (SMA) is caused by deficient expression of survival motor neuron (SMN) protein. New SMN-enhancing therapeutics are associated with variable clinical benefits. Limited knowledge of baseline and drug-induced SMN levels in disease-relevant tissues hinders efforts to optimize these treatments.METHODSSMN mRNA and protein levels were quantified in human tissues isolated during expedited autopsies.RESULTSSMN protein expression varied broadly among prenatal control spinal cord samples, but was restricted at relatively low levels in controls and SMA patients after 3 months of life. A 2.3-fold perinatal decrease in median SMN protein levels was not paralleled by comparable changes in SMN mRNA. In tissues isolated from nusinersen-treated SMA patients, antisense oligonucleotide (ASO) concentration and full-length (exon 7 including) SMN2 (SMN2-FL) mRNA level increases were highest in lumbar and thoracic spinal cord. An increased number of cells showed SMN immunolabeling in spinal cord of treated patients, but was not associated with an increase in whole-tissue SMN protein levels.CONCLUSIONSA normally occurring perinatal decrease in whole-tissue SMN protein levels supports efforts to initiate SMN-inducing therapies as soon after birth as possible. Limited ASO distribution to rostral spinal and brain regions in some patients likely limits clinical response of motor units in these regions for those patients. These results have important implications for optimizing treatment of SMA patients and warrant further investigations to enhance bioavailability of intrathecally administered ASOs.FUNDINGSMA Foundation, SMART, NIH (R01-NS096770, R01-NS062869), Ionis Pharmaceuticals, and PTC Therapeutics. Biogen provided support for absolute real-time RT-PCR.
    MeSH term(s) Aging/genetics ; Aging/metabolism ; Aging/pathology ; Autopsy ; Cell Survival ; Female ; Humans ; Male ; Motor Neurons/metabolism ; Motor Neurons/pathology ; Muscular Atrophy, Spinal/drug therapy ; Muscular Atrophy, Spinal/genetics ; Muscular Atrophy, Spinal/metabolism ; Muscular Atrophy, Spinal/pathology ; Oligodeoxyribonucleotides, Antisense/administration & dosage ; Spinal Cord/metabolism ; Spinal Cord/pathology ; Survival of Motor Neuron 2 Protein/antagonists & inhibitors ; Survival of Motor Neuron 2 Protein/genetics ; Survival of Motor Neuron 2 Protein/metabolism
    Chemical Substances Oligodeoxyribonucleotides, Antisense ; SMN2 protein, human ; Survival of Motor Neuron 2 Protein
    Language English
    Publishing date 2019-10-07
    Publishing country United States
    Document type Clinical Trial ; Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 3067-3
    ISSN 1558-8238 ; 0021-9738
    ISSN (online) 1558-8238
    ISSN 0021-9738
    DOI 10.1172/JCI124120
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: ELP1 Splicing Correction Reverses Proprioceptive Sensory Loss in Familial Dysautonomia.

    Morini, Elisabetta / Gao, Dadi / Montgomery, Connor M / Salani, Monica / Mazzasette, Chiara / Krussig, Tobias A / Swain, Brooke / Dietrich, Paula / Narasimhan, Jana / Gabbeta, Vijayalakshmi / Dakka, Amal / Hedrick, Jean / Zhao, Xin / Weetall, Marla / Naryshkin, Nikolai A / Wojtkiewicz, Gregory G / Ko, Chien-Ping / Talkowski, Michael E / Dragatsis, Ioannis /
    Slaugenhaupt, Susan A

    American journal of human genetics

    2019  Volume 104, Issue 4, Page(s) 638–650

    Abstract: Familial dysautonomia (FD) is a recessive neurodegenerative disease caused by a splice mutation in Elongator complex protein 1 (ELP1, also known as IKBKAP); this mutation leads to variable skipping of exon 20 and to a drastic reduction of ELP1 in the ... ...

    Abstract Familial dysautonomia (FD) is a recessive neurodegenerative disease caused by a splice mutation in Elongator complex protein 1 (ELP1, also known as IKBKAP); this mutation leads to variable skipping of exon 20 and to a drastic reduction of ELP1 in the nervous system. Clinically, many of the debilitating aspects of the disease are related to a progressive loss of proprioception; this loss leads to severe gait ataxia, spinal deformities, and respiratory insufficiency due to neuromuscular incoordination. There is currently no effective treatment for FD, and the disease is ultimately fatal. The development of a drug that targets the underlying molecular defect provides hope that the drastic peripheral neurodegeneration characteristic of FD can be halted. We demonstrate herein that the FD mouse TgFD9;Ikbkap
    MeSH term(s) Alleles ; Animals ; Behavior, Animal ; Cell Line ; Crosses, Genetic ; Disease Models, Animal ; Dysautonomia, Familial/genetics ; Dysautonomia, Familial/therapy ; Exons ; Fibroblasts ; Genotype ; Humans ; Introns ; Kinetin/genetics ; Kinetin/therapeutic use ; Male ; Mice ; Mice, Inbred C57BL ; Mutation ; Neurons/metabolism ; Phenotype ; Proprioception ; RNA Splicing ; Transcriptional Elongation Factors/genetics
    Chemical Substances Elp1 protein, human ; Transcriptional Elongation Factors ; Kinetin (P39Y9652YJ)
    Language English
    Publishing date 2019-03-21
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 219384-x
    ISSN 1537-6605 ; 0002-9297
    ISSN (online) 1537-6605
    ISSN 0002-9297
    DOI 10.1016/j.ajhg.2019.02.009
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: Discovery and Optimization of Small Molecule Splicing Modifiers of Survival Motor Neuron 2 as a Treatment for Spinal Muscular Atrophy.

    Woll, Matthew G / Qi, Hongyan / Turpoff, Anthony / Zhang, Nanjing / Zhang, Xiaoyan / Chen, Guangming / Li, Chunshi / Huang, Song / Yang, Tianle / Moon, Young-Choon / Lee, Chang-Sun / Choi, Soongyu / Almstead, Neil G / Naryshkin, Nikolai A / Dakka, Amal / Narasimhan, Jana / Gabbeta, Vijayalakshmi / Welch, Ellen / Zhao, Xin /
    Risher, Nicole / Sheedy, Josephine / Weetall, Marla / Karp, Gary M

    Journal of medicinal chemistry

    2016  Volume 59, Issue 13, Page(s) 6070–6085

    Abstract: The underlying cause of spinal muscular atrophy (SMA) is a deficiency of the survival motor neuron (SMN) protein. Starting from hits identified in a high-throughput screening campaign and through structure-activity relationship investigations, we have ... ...

    Abstract The underlying cause of spinal muscular atrophy (SMA) is a deficiency of the survival motor neuron (SMN) protein. Starting from hits identified in a high-throughput screening campaign and through structure-activity relationship investigations, we have developed small molecules that potently shift the alternative splicing of the SMN2 exon 7, resulting in increased production of the full-length SMN mRNA and protein. Three novel chemical series, represented by compounds 9, 14, and 20, have been optimized to increase the level of SMN protein by >50% in SMA patient-derived fibroblasts at concentrations of <160 nM. Daily administration of these compounds to severe SMA Δ7 mice results in an increased production of SMN protein in disease-relevant tissues and a significant increase in median survival time in a dose-dependent manner. Our work supports the development of an orally administered small molecule for the treatment of patients with SMA.
    MeSH term(s) Alternative Splicing/drug effects ; Animals ; Cell Line ; Drug Discovery ; Exons/drug effects ; HEK293 Cells ; Humans ; Mice, Knockout ; Muscular Atrophy, Spinal/drug therapy ; Muscular Atrophy, Spinal/genetics ; RNA, Messenger/genetics ; Small Molecule Libraries/administration & dosage ; Small Molecule Libraries/chemistry ; Small Molecule Libraries/pharmacology ; Small Molecule Libraries/therapeutic use ; Structure-Activity Relationship ; Survival of Motor Neuron 2 Protein/genetics
    Chemical Substances RNA, Messenger ; SMN2 protein, human ; Small Molecule Libraries ; Survival of Motor Neuron 2 Protein
    Language English
    Publishing date 2016-07-14
    Publishing country United States
    Document type Journal Article
    ZDB-ID 218133-2
    ISSN 1520-4804 ; 0022-2623
    ISSN (online) 1520-4804
    ISSN 0022-2623
    DOI 10.1021/acs.jmedchem.6b00460
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: Motor neuron disease. SMN2 splicing modifiers improve motor function and longevity in mice with spinal muscular atrophy.

    Naryshkin, Nikolai A / Weetall, Marla / Dakka, Amal / Narasimhan, Jana / Zhao, Xin / Feng, Zhihua / Ling, Karen K Y / Karp, Gary M / Qi, Hongyan / Woll, Matthew G / Chen, Guangming / Zhang, Nanjing / Gabbeta, Vijayalakshmi / Vazirani, Priya / Bhattacharyya, Anuradha / Furia, Bansri / Risher, Nicole / Sheedy, Josephine / Kong, Ronald /
    Ma, Jiyuan / Turpoff, Anthony / Lee, Chang-Sun / Zhang, Xiaoyan / Moon, Young-Choon / Trifillis, Panayiota / Welch, Ellen M / Colacino, Joseph M / Babiak, John / Almstead, Neil G / Peltz, Stuart W / Eng, Loren A / Chen, Karen S / Mull, Jesse L / Lynes, Maureen S / Rubin, Lee L / Fontoura, Paulo / Santarelli, Luca / Haehnke, Daniel / McCarthy, Kathleen D / Schmucki, Roland / Ebeling, Martin / Sivaramakrishnan, Manaswini / Ko, Chien-Ping / Paushkin, Sergey V / Ratni, Hasane / Gerlach, Irene / Ghosh, Anirvan / Metzger, Friedrich

    Science (New York, N.Y.)

    2014  Volume 345, Issue 6197, Page(s) 688–693

    Abstract: Spinal muscular atrophy (SMA) is a genetic disease caused by mutation or deletion of the survival of motor neuron 1 (SMN1) gene. A paralogous gene in humans, SMN2, produces low, insufficient levels of functional SMN protein due to alternative splicing ... ...

    Abstract Spinal muscular atrophy (SMA) is a genetic disease caused by mutation or deletion of the survival of motor neuron 1 (SMN1) gene. A paralogous gene in humans, SMN2, produces low, insufficient levels of functional SMN protein due to alternative splicing that truncates the transcript. The decreased levels of SMN protein lead to progressive neuromuscular degeneration and high rates of mortality. Through chemical screening and optimization, we identified orally available small molecules that shift the balance of SMN2 splicing toward the production of full-length SMN2 messenger RNA with high selectivity. Administration of these compounds to Δ7 mice, a model of severe SMA, led to an increase in SMN protein levels, improvement of motor function, and protection of the neuromuscular circuit. These compounds also extended the life span of the mice. Selective SMN2 splicing modifiers may have therapeutic potential for patients with SMA.
    MeSH term(s) Administration, Oral ; Alternative Splicing/drug effects ; Animals ; Cells, Cultured ; Coumarins/administration & dosage ; Coumarins/chemistry ; Disease Models, Animal ; Drug Evaluation, Preclinical ; Humans ; Isocoumarins/administration & dosage ; Isocoumarins/chemistry ; Longevity/drug effects ; Mice ; Muscular Atrophy, Spinal/drug therapy ; Muscular Atrophy, Spinal/genetics ; Muscular Atrophy, Spinal/metabolism ; Pyrimidinones/administration & dosage ; Pyrimidinones/chemistry ; RNA, Messenger/genetics ; Sequence Deletion ; Small Molecule Libraries/administration & dosage ; Small Molecule Libraries/chemistry ; Survival of Motor Neuron 2 Protein/genetics ; Survival of Motor Neuron 2 Protein/metabolism
    Chemical Substances Coumarins ; Isocoumarins ; Pyrimidinones ; RNA, Messenger ; Small Molecule Libraries ; Survival of Motor Neuron 2 Protein
    Language English
    Publishing date 2014-08-08
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 128410-1
    ISSN 1095-9203 ; 0036-8075
    ISSN (online) 1095-9203
    ISSN 0036-8075
    DOI 10.1126/science.1250127
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top