LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 51

Search options

  1. Article ; Online: Outcome Measures in Facioscapulohumeral Muscular Dystrophy Clinical Trials.

    Ghasemi, Mehdi / Emerson, Charles P / Hayward, Lawrence J

    Cells

    2022  Volume 11, Issue 4

    Abstract: Facioscapulohumeral muscular dystrophy (FSHD) is a debilitating muscular dystrophy with a variable age of onset, severity, and progression. While there is still no cure for this disease, progress towards FSHD therapies has accelerated since the ... ...

    Abstract Facioscapulohumeral muscular dystrophy (FSHD) is a debilitating muscular dystrophy with a variable age of onset, severity, and progression. While there is still no cure for this disease, progress towards FSHD therapies has accelerated since the underlying mechanism of epigenetic derepression of the double homeobox 4 (
    MeSH term(s) Homeodomain Proteins/metabolism ; Humans ; Muscle, Skeletal/metabolism ; Muscular Dystrophy, Facioscapulohumeral/drug therapy ; Muscular Dystrophy, Facioscapulohumeral/therapy ; Outcome Assessment, Health Care
    Chemical Substances Homeodomain Proteins
    Language English
    Publishing date 2022-02-16
    Publishing country Switzerland
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't ; Review
    ZDB-ID 2661518-6
    ISSN 2073-4409 ; 2073-4409
    ISSN (online) 2073-4409
    ISSN 2073-4409
    DOI 10.3390/cells11040687
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: Lower Ca2+ enhances the K+-induced force depression in normal and HyperKPP mouse muscles.

    Uwera, Francine / Ammar, Tarek / McRae, Callum / Hayward, Lawrence J / Renaud, Jean-Marc

    The Journal of general physiology

    2020  Volume 152, Issue 7

    Abstract: Hyperkalemic periodic paralysis (HyperKPP) manifests as stiffness or subclinical myotonic discharges before or during periods of episodic muscle weakness or paralysis. Ingestion of Ca2+ alleviates HyperKPP symptoms, but the mechanism is unknown because ... ...

    Abstract Hyperkalemic periodic paralysis (HyperKPP) manifests as stiffness or subclinical myotonic discharges before or during periods of episodic muscle weakness or paralysis. Ingestion of Ca2+ alleviates HyperKPP symptoms, but the mechanism is unknown because lowering extracellular [Ca2+] ([Ca2+]e) has no effect on force development in normal muscles under normal conditions. Lowering [Ca2+]e, however, is known to increase the inactivation of voltage-gated cation channels, especially when the membrane is depolarized. Two hypotheses were tested: (1) lowering [Ca2+]e depresses force in normal muscles under conditions that depolarize the cell membrane; and (2) HyperKPP muscles have a greater sensitivity to low Ca2+-induced force depression because many fibers are depolarized, even at a normal [K+]e. In wild type muscles, lowering [Ca2+]e from 2.4 to 0.3 mM had little effect on tetanic force and membrane excitability at a normal K+ concentration of 4.7 mM, whereas it significantly enhanced K+-induced depression of force and membrane excitability. In HyperKPP muscles, lowering [Ca2+]e enhanced the K+-induced loss of force and membrane excitability not only at elevated [K+]e but also at 4.7 mM K+. Lowering [Ca2+]e increased the incidence of generating fast and transient contractures and gave rise to a slower increase in unstimulated force, especially in HyperKPP muscles. Lowering [Ca2+]e reduced the efficacy of salbutamol, a β2 adrenergic receptor agonist and a treatment for HyperKPP, to increase force at elevated [K+]e. Replacing Ca2+ by an equivalent concentration of Mg2+ neither fully nor consistently reverses the effects of lowering [Ca2+]e. These results suggest that the greater Ca2+ sensitivity of HyperKPP muscles primarily relates to (1) a greater effect of Ca2+ in depolarized fibers and (2) an increased proportion of depolarized HyperKPP muscle fibers compared with control muscle fibers, even at normal [K+]e.
    MeSH term(s) Animals ; Calcium/metabolism ; Mice ; Muscle Contraction ; Muscle Fibers, Skeletal/physiology ; Muscle, Skeletal/physiology ; Muscle, Skeletal/physiopathology ; Paralysis, Hyperkalemic Periodic ; Potassium/metabolism
    Chemical Substances Potassium (RWP5GA015D) ; Calcium (SY7Q814VUP)
    Language English
    Publishing date 2020-04-15
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 3118-5
    ISSN 1540-7748 ; 0022-1295
    ISSN (online) 1540-7748
    ISSN 0022-1295
    DOI 10.1085/jgp.201912511
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: Renewal of oligodendrocyte lineage reverses dysmyelination and CNS neurodegeneration through corrected N-acetylaspartate metabolism.

    Lotun, Anoushka / Li, Danning / Xu, Hongxia / Su, Qin / Tuncer, Serafettin / Sanmiguel, Julio / Mooney, Morgan / Baer, Christina E / Ulbrich, Russell / Eyles, Stephen J / Strittmatter, Lara / Hayward, Lawrence J / Gessler, Dominic J / Gao, Guangping

    Progress in neurobiology

    2023  Volume 226, Page(s) 102460

    Abstract: Myelinating oligodendrocytes are essential for neuronal communication and homeostasis of the central nervous system (CNS). One of the most abundant molecules in the mammalian CNS is N-acetylaspartate (NAA), which is catabolized into L-aspartate and ... ...

    Abstract Myelinating oligodendrocytes are essential for neuronal communication and homeostasis of the central nervous system (CNS). One of the most abundant molecules in the mammalian CNS is N-acetylaspartate (NAA), which is catabolized into L-aspartate and acetate by the enzyme aspartoacylase (ASPA) in oligodendrocytes. The resulting acetate moiety is thought to contribute to myelin lipid synthesis. In addition, affected NAA metabolism has been implicated in several neurological disorders, including leukodystrophies and demyelinating diseases such as multiple sclerosis. Genetic disruption of ASPA function causes Canavan disease, which is hallmarked by increased NAA levels, myelin and neuronal loss, large vacuole formation in the CNS, and early death in childhood. Although NAA's direct role in the CNS is inconclusive, in peripheral adipose tissue, NAA-derived acetate has been found to modify histones, a mechanism known to be involved in epigenetic regulation of cell differentiation. We hypothesize that a lack of cellular differentiation in the brain contributes to the disruption of myelination and neurodegeneration in diseases with altered NAA metabolism, such as Canavan disease. Our study demonstrates that loss of functional Aspa in mice disrupts myelination and shifts the transcriptional expression of neuronal and oligodendrocyte markers towards less differentiated stages in a spatiotemporal manner. Upon re-expression of ASPA, these oligodendrocyte and neuronal lineage markers are either improved or normalized, suggesting that NAA breakdown by Aspa plays an essential role in the maturation of neurons and oligodendrocytes. Also, this effect of ASPA re-expression is blunted in old mice, potentially due to limited ability of neuronal, rather than oligodendrocyte, recovery.
    MeSH term(s) Mice ; Animals ; Canavan Disease/genetics ; Canavan Disease/metabolism ; Cell Lineage ; Epigenesis, Genetic ; Central Nervous System/metabolism ; Oligodendroglia ; Myelin Sheath/metabolism ; Mammals
    Chemical Substances N-acetylaspartate (997-55-7)
    Language English
    Publishing date 2023-05-04
    Publishing country England
    Document type Journal Article
    ZDB-ID 185535-9
    ISSN 1873-5118 ; 0301-0082
    ISSN (online) 1873-5118
    ISSN 0301-0082
    DOI 10.1016/j.pneurobio.2023.102460
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Lessons learned from muscle fatigue: implications for treatment of patients with hyperkalemic periodic paralysis.

    Renaud, Jean-Marc / Hayward, Lawrence J

    Recent patents on biotechnology

    2012  Volume 6, Issue 3, Page(s) 184–191

    Abstract: Hyperkalemic periodic paralysis (HyperKPP) is a disease characterized by periods of myotonic discharges and paralytic attacks causing weakness, the latter associated with increases in plasma [K(+)]. The myotonic discharge is due to increased Na(+) influx ...

    Abstract Hyperkalemic periodic paralysis (HyperKPP) is a disease characterized by periods of myotonic discharges and paralytic attacks causing weakness, the latter associated with increases in plasma [K(+)]. The myotonic discharge is due to increased Na(+) influx through defective Na(+) channels that triggers generation of several action potentials. The subsequent increase in extracellular K(+) concentration causes excessive membrane depolarization that inactivates Na(+) channels triggering the paralysis. None of the available treatments is fully effective. This paper reviews the capacity of Na(+) K(+)ATPase pumps, KATP and ClC-1 Cl(-) channels in improving membrane excitability during muscle activity and how using these three membrane components we can study future and more effective treatments for HyperKPP patients. The review of current patents related to HyperKPP reinforces the need of novel approaches for the treatment of this channelopathy.
    MeSH term(s) Humans ; Muscle Fatigue/physiology ; Mutation, Missense/genetics ; Paralysis, Hyperkalemic Periodic/physiopathology ; Paralysis, Hyperkalemic Periodic/therapy ; Patents as Topic ; Potassium Channels/metabolism ; Sodium Channels/genetics
    Chemical Substances Potassium Channels ; Sodium Channels
    Language English
    Publishing date 2012-10-23
    Publishing country United Arab Emirates
    Document type Journal Article ; Review
    ISSN 2212-4012
    ISSN (online) 2212-4012
    DOI 10.2174/1872208311206030184
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: Meeting report: the 2021 FSHD International Research Congress.

    Jagannathan, Sujatha / de Greef, Jessica C / Hayward, Lawrence J / Yokomori, Kyoko / Gabellini, Davide / Mul, Karlien / Sacconi, Sabrina / Arjomand, Jamshid / Kinoshita, June / Harper, Scott Q

    Skeletal muscle

    2022  Volume 12, Issue 1, Page(s) 1

    Abstract: Facioscapulohumeral muscular dystrophy (FSHD) is the second most common genetic myopathy, characterized by slowly progressing and highly heterogeneous muscle wasting with a typical onset in the late teens/early adulthood [1]. Although the etiology of the ...

    Abstract Facioscapulohumeral muscular dystrophy (FSHD) is the second most common genetic myopathy, characterized by slowly progressing and highly heterogeneous muscle wasting with a typical onset in the late teens/early adulthood [1]. Although the etiology of the disease for both FSHD type 1 and type 2 has been attributed to gain-of-toxic function stemming from aberrant DUX4 expression, the exact pathogenic mechanisms involved in muscle wasting have yet to be elucidated [2-4]. The 2021 FSHD International Research Congress, held virtually on June 24-25, convened over 350 researchers and clinicians to share the most recent advances in the understanding of the disease mechanism, discuss the proliferation of interventional strategies and refinement of clinical outcome measures, including results from the ReDUX4 trial, a phase 2b clinical trial of losmapimod in FSHD [NCT04003974].
    MeSH term(s) Adolescent ; Adult ; Homeodomain Proteins/genetics ; Humans ; Muscle, Skeletal/metabolism ; Muscular Atrophy/metabolism ; Muscular Dystrophy, Facioscapulohumeral/metabolism
    Chemical Substances Homeodomain Proteins
    Language English
    Publishing date 2022-01-17
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't ; Review
    ZDB-ID 2595637-1
    ISSN 2044-5040 ; 2044-5040
    ISSN (online) 2044-5040
    ISSN 2044-5040
    DOI 10.1186/s13395-022-00287-8
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: iMyoblasts for ex vivo and in vivo investigations of human myogenesis and disease modeling.

    Guo, Dongsheng / Daman, Katelyn / Chen, Jennifer Jc / Shi, Meng-Jiao / Yan, Jing / Matijasevic, Zdenka / Rickard, Amanda M / Bennett, Monica H / Kiselyov, Alex / Zhou, Haowen / Bang, Anne G / Wagner, Kathryn R / Maehr, René / King, Oliver D / Hayward, Lawrence J / Emerson, Charles P

    eLife

    2022  Volume 11

    Abstract: Skeletal muscle myoblasts (iMyoblasts) were generated from human induced pluripotent stem cells (iPSCs) using an efficient and reliable transgene-free induction and stem cell selection protocol. Immunofluorescence, flow cytometry, qPCR, digital RNA ... ...

    Abstract Skeletal muscle myoblasts (iMyoblasts) were generated from human induced pluripotent stem cells (iPSCs) using an efficient and reliable transgene-free induction and stem cell selection protocol. Immunofluorescence, flow cytometry, qPCR, digital RNA expression profiling, and scRNA-Seq studies identify iMyoblasts as a
    MeSH term(s) Animals ; Cell Differentiation ; Cell Line ; Cell Lineage ; Cells, Cultured ; Disease Models, Animal ; Homeodomain Proteins/metabolism ; Humans ; Induced Pluripotent Stem Cells/cytology ; Induced Pluripotent Stem Cells/transplantation ; Mice ; Muscle Development ; Muscular Dystrophy, Facioscapulohumeral/pathology ; Muscular Dystrophy, Facioscapulohumeral/therapy ; Myoblasts/transplantation ; PAX3 Transcription Factor/metabolism ; Recovery of Function ; Regeneration
    Chemical Substances Dux4 protein, mouse ; Homeodomain Proteins ; PAX3 Transcription Factor ; Pax3 protein, mouse (138016-91-8)
    Language English
    Publishing date 2022-01-25
    Publishing country England
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 2687154-3
    ISSN 2050-084X ; 2050-084X
    ISSN (online) 2050-084X
    ISSN 2050-084X
    DOI 10.7554/eLife.70341
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: Understanding the physiology of the asymptomatic diaphragm of the M1592V hyperkalemic periodic paralysis mouse.

    Ammar, Tarek / Lin, Wei / Higgins, Amanda / Hayward, Lawrence J / Renaud, Jean-Marc

    The Journal of general physiology

    2015  Volume 146, Issue 6, Page(s) 509–525

    Abstract: The diaphragm muscle of hyperkalemic periodic paralysis (HyperKPP) patients and of the M1592V HyperKPP mouse model rarely suffers from the myotonic and paralytic symptoms that occur in limb muscles. Enigmatically, HyperKPP diaphragm expresses the mutant ... ...

    Abstract The diaphragm muscle of hyperkalemic periodic paralysis (HyperKPP) patients and of the M1592V HyperKPP mouse model rarely suffers from the myotonic and paralytic symptoms that occur in limb muscles. Enigmatically, HyperKPP diaphragm expresses the mutant NaV1.4 channel and, more importantly, has an abnormally high Na(+) influx similar to that in extensor digitorum longus (EDL) and soleus, two hindlimb muscles suffering from the robust HyperKPP abnormalities. The objective was to uncover the physiological mechanisms that render HyperKPP diaphragm asymptomatic. A first mechanism involves efficient maintenance of resting membrane polarization in HyperKPP diaphragm at various extracellular K(+) concentrations compared with larger membrane depolarizations in HyperKPP EDL and soleus. The improved resting membrane potential (EM) results from significantly increased Na(+) K(+) pump electrogenic activity, and not from an increased protein content. Action potential amplitude was greater in HyperKPP diaphragm than in HyperKPP soleus and EDL, providing a second mechanism for the asymptomatic behavior of the HyperKPP diaphragm. One suggested mechanism for the greater action potential amplitude is lower intracellular Na(+) concentration because of greater Na(+) K(+) pump activity, allowing better Na(+) current during the action potential depolarization phase. Finally, HyperKPP diaphragm had a greater capacity to generate force at depolarized EM compared with wild-type diaphragm. Action potential amplitude was not different between wild-type and HyperKPP diaphragm. There was also no evidence for an increased activity of the Na(+)-Ca(2+) exchanger working in the reverse mode in the HyperKPP diaphragm compared with the wild-type diaphragm. So, a third mechanism remains to be elucidated to fully understand how HyperKPP diaphragm generates more force compared with wild type. Although the mechanism for the greater force at depolarized resting EM remains to be determined, this study provides support for the modulation of the Na(+) K(+) pump as a component of therapy to alleviate weakness in HyperKPP.
    MeSH term(s) Action Potentials ; Animals ; Diaphragm/drug effects ; Diaphragm/metabolism ; Diaphragm/physiopathology ; Membrane Potentials ; Mice ; Mutation, Missense ; NAV1.4 Voltage-Gated Sodium Channel/genetics ; NAV1.4 Voltage-Gated Sodium Channel/metabolism ; Paralysis, Hyperkalemic Periodic/genetics ; Paralysis, Hyperkalemic Periodic/metabolism ; Paralysis, Hyperkalemic Periodic/physiopathology ; Potassium/metabolism ; Potassium/pharmacology ; Sodium/metabolism
    Chemical Substances NAV1.4 Voltage-Gated Sodium Channel ; Scn4a protein, mouse ; Sodium (9NEZ333N27) ; Potassium (RWP5GA015D)
    Language English
    Publishing date 2015-12
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 3118-5
    ISSN 1540-7748 ; 0022-1295
    ISSN (online) 1540-7748
    ISSN 0022-1295
    DOI 10.1085/jgp.201511476
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article: Mutant SOD1 instability: implications for toxicity in amyotrophic lateral sclerosis.

    Tiwari, Ashutosh / Hayward, Lawrence J

    Neuro-degenerative diseases

    2005  Volume 2, Issue 3-4, Page(s) 115–127

    Abstract: The biological basis of preferential motor neuron degeneration in amyotrophic lateral sclerosis (ALS) remains incompletely understood, and effective therapies to prevent the lethal consequences of this disorder are not yet available. Since 1993, more ... ...

    Abstract The biological basis of preferential motor neuron degeneration in amyotrophic lateral sclerosis (ALS) remains incompletely understood, and effective therapies to prevent the lethal consequences of this disorder are not yet available. Since 1993, more than 100 mutant variants of the antioxidant enzyme Cu/Zn superoxide dismutase (SOD1) have been identified in familial ALS. Many studies have sought to distinguish abnormal properties shared by these proteins that may contribute to their toxic effects and cause age-dependent motor neuron loss. Complex networks of cellular interactions and changes associated with aging may link mutant SOD1s and other stresses to motor neuron death in ALS. Our laboratory and collaborators have compared physicochemical properties of biologically metallated wild-type and mutant SOD1 proteins to discern specific vulnerabilities that may be relevant to the mutant toxicity in vivo. X-ray crystal structures obtained from metallated 'wild-type-like' (WTL) SOD1 mutants, which retain the ability to bind copper and zinc and exhibit normal specific activity, indicate a native-like structure with only subtle changes to the backbone fold. In contrast, a group of 'metal-binding region' (MBR) SOD1 mutants that are deficient in copper and zinc exhibit severe thermal destabilization and structural disorder of conserved loops near the metal-binding sites. A growing body of evidence highlights specific stresses in vivo that may perturb well-folded, metallated SOD1 variants and thereby favor an increased burden of partially unfolded, metal-deficient species. For example, WTL SOD1 mutants are more susceptible than wild-type SOD1 to reduction of the intrasubunit disulfide bond between Cys-57 and Cys-146 at physiological pH and temperature. This bond anchors the disulfide loop to the SOD1 beta-barrel and helps to maintain the dimeric configuration of the protein. Cleavage of the disulfide linkage renders the well-folded WTL mutants vulnerable to metal loss and monomerization such that they may resemble the destabilized and locally misfolded MBR mutant species. SOD1 proteins with disordered loops or monomeric structure are expected to be more susceptible to aberrant self-association or detrimental interactions with other cellular constituents. The challenge for future investigations is to relate these abnormal properties of partially unfolded SOD1 to specific mechanisms of toxicity in motor neurons, supporting cells, or target tissues.
    MeSH term(s) Amyotrophic Lateral Sclerosis/enzymology ; Animals ; Humans ; Mutation ; Protein Structure, Quaternary ; Superoxide Dismutase/chemistry ; Superoxide Dismutase/metabolism ; Superoxide Dismutase/toxicity ; Superoxide Dismutase-1
    Chemical Substances SOD1 protein, human ; Superoxide Dismutase (EC 1.15.1.1) ; Superoxide Dismutase-1 (EC 1.15.1.1)
    Language English
    Publishing date 2005
    Publishing country Switzerland
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't ; Review
    ZDB-ID 2143569-8
    ISSN 1660-2862 ; 1660-2854
    ISSN (online) 1660-2862
    ISSN 1660-2854
    DOI 10.1159/000089616
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article ; Online: Safety and efficacy of losmapimod in facioscapulohumeral muscular dystrophy (ReDUX4): a randomised, double-blind, placebo-controlled phase 2b trial.

    Tawil, Rabi / Wagner, Kathryn R / Hamel, Johanna I / Leung, Doris G / Statland, Jeffrey M / Wang, Leo H / Genge, Angela / Sacconi, Sabrina / Lochmüller, Hanns / Reyes-Leiva, David / Diaz-Manera, Jordi / Alonso-Perez, Jorge / Muelas, Nuria / Vilchez, Juan J / Pestronk, Alan / Gibson, Summer / Goyal, Namita A / Hayward, Lawrence J / Johnson, Nicholas /
    LoRusso, Samantha / Freimer, Miriam / Shieh, Perry B / Subramony, S H / van Engelen, Baziel / Kools, Joost / Leinhard, Olof Dahlqvist / Widholm, Per / Morabito, Christopher / Moxham, Christopher M / Cadavid, Diego / Mellion, Michelle L / Odueyungbo, Adefowope / Tracewell, William G / Accorsi, Anthony / Ronco, Lucienne / Gould, Robert J / Shoskes, Jennifer / Rojas, Luis Alejandro / Jiang, John G

    The Lancet. Neurology

    2024  Volume 23, Issue 5, Page(s) 477–486

    Abstract: Background: Facioscapulohumeral muscular dystrophy is a hereditary progressive myopathy caused by aberrant expression of the transcription factor DUX4 in skeletal muscle. No approved disease-modifying treatments are available for this disorder. We aimed ...

    Abstract Background: Facioscapulohumeral muscular dystrophy is a hereditary progressive myopathy caused by aberrant expression of the transcription factor DUX4 in skeletal muscle. No approved disease-modifying treatments are available for this disorder. We aimed to assess the safety and efficacy of losmapimod (a small molecule that inhibits p38α MAPK, a regulator of DUX4 expression, and p38β MAPK) for the treatment of facioscapulohumeral muscular dystrophy.
    Methods: We did a randomised, double-blind, placebo-controlled phase 2b trial at 17 neurology centres in Canada, France, Spain, and the USA. We included adults aged 18-65 years with type 1 facioscapulohumeral muscular dystrophy (ie, with loss of repression of DUX4 expression, as ascertained by genotyping), a Ricci clinical severity score of 2-4, and at least one skeletal muscle judged using MRI to be suitable for biopsy. Participants were randomly allocated (1:1) to either oral losmapimod (15 mg twice a day) or matching placebo for 48 weeks, via an interactive response technology system. The investigator, study staff, participants, sponsor, primary outcome assessors, and study monitor were masked to the treatment allocation until study closure. The primary endpoint was change from baseline to either week 16 or 36 in DUX4-driven gene expression in skeletal muscle biopsy samples, as measured by quantitative RT-PCR. The primary efficacy analysis was done in all participants who were randomly assigned and who had available data for assessment, according to the modified intention-to-treat principle. Safety and tolerability were assessed as secondary endpoints. This study is registered at ClinicalTrials.gov, number NCT04003974. The phase 2b trial is complete; an open-label extension is ongoing.
    Findings: Between Aug 27, 2019, and Feb 27, 2020, 80 people were enrolled. 40 were randomly allocated to losmapimod and 40 to placebo. 54 (68%) participants were male and 26 (33%) were female, 70 (88%) were White, and mean age was 45·7 (SD 12·5) years. Least squares mean changes from baseline in DUX4-driven gene expression did not differ significantly between the losmapimod (0·83 [SE 0·61]) and placebo (0·40 [0·65]) groups (difference 0·43 [SE 0·56; 95% CI -1·04 to 1·89]; p=0·56). Losmapimod was well tolerated. 29 treatment-emergent adverse events (nine drug-related) were reported in the losmapimod group compared with 23 (two drug-related) in the placebo group. Two participants in the losmapimod group had serious adverse events that were deemed unrelated to losmapimod by the investigators (alcohol poisoning and suicide attempt; postoperative wound infection) compared with none in the placebo group. No treatment discontinuations due to adverse events occurred and no participants died during the study.
    Interpretation: Although losmapimod did not significantly change DUX4-driven gene expression, it was associated with potential improvements in prespecified structural outcomes (muscle fat infiltration), functional outcomes (reachable workspace, a measure of shoulder girdle function), and patient-reported global impression of change compared with placebo. These findings have informed the design and choice of efficacy endpoints for a phase 3 study of losmapimod in adults with facioscapulohumeral muscular dystrophy.
    Funding: Fulcrum Therapeutics.
    MeSH term(s) Adult ; Humans ; Male ; Female ; Middle Aged ; Muscular Dystrophy, Facioscapulohumeral ; Treatment Outcome ; Pyridines ; Cyclopropanes ; Double-Blind Method
    Chemical Substances 6-(5-((cyclopropylamino)carbonyl)-3-fluoro-2-methylphenyl)-N-(2,2-dimethylprpyl)-3-pyridinecarboxamide ; Pyridines ; Cyclopropanes
    Language English
    Publishing date 2024-04-17
    Publishing country England
    Document type Randomized Controlled Trial ; Journal Article
    ZDB-ID 2081241-3
    ISSN 1474-4465 ; 1474-4422
    ISSN (online) 1474-4465
    ISSN 1474-4422
    DOI 10.1016/S1474-4422(24)00073-5
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article: Familial amyotrophic lateral sclerosis mutants of copper/zinc superoxide dismutase are susceptible to disulfide reduction.

    Tiwari, Ashutosh / Hayward, Lawrence J

    The Journal of biological chemistry

    2002  Volume 278, Issue 8, Page(s) 5984–5992

    Abstract: We observed that 14 biologically metallated mutants of copper/zinc superoxide dismutase (SOD1) associated with familial amyotrophic lateral sclerosis all exhibited aberrantly accelerated mobility during partially denaturing PAGE and increased sensitivity ...

    Abstract We observed that 14 biologically metallated mutants of copper/zinc superoxide dismutase (SOD1) associated with familial amyotrophic lateral sclerosis all exhibited aberrantly accelerated mobility during partially denaturing PAGE and increased sensitivity to proteolytic digestion compared with wild type SOD1. Decreased metal binding site occupancy and exposure to the disulfide-reducing agents dithiothreitol, Tris(2-carboxyethyl)phosphine (TCEP), or reduced glutathione increased the fraction of anomalously migrating mutant SOD1 proteins. Furthermore, the incubation of mutant SOD1s with TCEP increased the accessibility to iodoacetamide of cysteine residues that normally participate in the formation of the intrasubunit disulfide bond (Cys-57 to Cys-146) or are buried within the core of the beta-barrel (Cys-6). SOD1 enzymes in spinal cord lysates from G85R and G93A mutant but not wild type SOD1 transgenic mice also exhibited abnormal vulnerability to TCEP, which exposed normally inaccessible cysteine residues to modification by maleimide conjugated to polyethylene glycol. These results implicate SOD1 destabilization under cellular disulfide-reducing conditions at physiological pH and temperature as a shared property that may be relevant to amyotrophic lateral sclerosis mutant neurotoxicity.
    MeSH term(s) Amino Acid Substitution ; Animals ; Binding Sites ; Cysteine ; Disulfides/pharmacology ; Dithiothreitol/pharmacology ; Humans ; Iodoacetamide/pharmacology ; Kinetics ; Mice ; Motor Neuron Disease/enzymology ; Motor Neuron Disease/genetics ; Mutagenesis, Site-Directed ; Mutation ; Organ Specificity ; Oxidation-Reduction ; Protein Structure, Secondary ; Recombinant Proteins/chemistry ; Recombinant Proteins/metabolism ; Superoxide Dismutase/chemistry ; Superoxide Dismutase/genetics ; Superoxide Dismutase/metabolism
    Chemical Substances Disulfides ; Recombinant Proteins ; Superoxide Dismutase (EC 1.15.1.1) ; Cysteine (K848JZ4886) ; Dithiothreitol (T8ID5YZU6Y) ; Iodoacetamide (ZRH8M27S79)
    Language English
    Publishing date 2002-11-27
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't ; Research Support, U.S. Gov't, P.H.S.
    ZDB-ID 2997-x
    ISSN 1083-351X ; 0021-9258
    ISSN (online) 1083-351X
    ISSN 0021-9258
    DOI 10.1074/jbc.M210419200
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top