pubmed_id
stringlengths
41
43
abstract
stringlengths
3
18.8k
http://www.ncbi.nlm.nih.gov/pubmed/23225386
1. Muscle Nerve. 2012 Dec;46(6):951-3. doi: 10.1002/mus.23529. Patient-identified disease burden in facioscapulohumeral muscular dystrophy. Johnson NE(1), Quinn C, Eastwood E, Tawil R, Heatwole CR. Author information: (1)Department of Neurology, University of Rochester, 601 Elmwood Avenue, P.O. Box 673, Rochester, New York 14642, USA. Nicholas_johnson@urmc.rochester.edu INTRODUCTION: The multitude of symptoms associated with facioscapulohumeral muscular dystrophy (FSHD) disease burden are of varying importance. The extent of these symptoms and their cumulative effect on the FSHD population is unknown. METHODS: We conducted interviews with adult FSHD patients to identify which symptoms have the greatest effect on their lives. Each interview was recorded, transcribed, coded, and analyzed using a qualitative framework technique, triangulation, and a three-investigator consensus approach. RESULTS: One thousand three hundred seventy-five quotes were obtained through 20 patient interviews. Two hundred fifty-one symptoms of importance were identified representing 14 themes of FSHD disease burden. Symptoms associated with mobility impairment, activity limitation, and social role limitation were most frequently mentioned by participants. CONCLUSIONS: There are multiple themes and symptoms, some previously underrecognized, that play a key role in FSHD disease burden. Copyright © 2012 Wiley Periodicals, Inc. DOI: 10.1002/mus.23529 PMCID: PMC4097080 PMID: 23225386 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/30414142
1. Methods Mol Biol. 2019;1890:29-40. doi: 10.1007/978-1-4939-8900-3_3. Identification of Transcription Factor-Binding Sites in the Mouse FOXO1 Promoter. Cardozo CP(1)(2). Author information: (1)Center for the Medical Consequences of Spinal Cord Injury, James J Peters Medical Center, Bronx, NY, USA. christopher.cardozo@mssm.edu. (2)Icahn School of Medicine at Mount Sinai, New York, NY, USA. christopher.cardozo@mssm.edu. One critical determinant of levels of gene expression is binding of transcription factors to cognate DNA sequences in promoter and enhancer regions of target genes. Transcription factors are DNA-binding proteins to which transcriptional co-regulators are bound, ultimately resulting in histone modifications that change chromatin structure to regulate transcription. Examples of transcription factors include hormone-activated transcription factors such as the glucocorticoid receptor, transcription factors regulated by cell surface receptors such as FOXO1 and Smad2/Smad3, and many others. Promoter regions typically contain multiple, diverse transcription factor-binding sites. Binding sites for cell-type-specific transcription factors involved in cell fate determination such as Runx2, MyoD, or myogenin are frequently observed. Promoter regions are located within ~2 kb upstream of the transcriptional start site, whereas enhancers may be located at some distance from promoter sequences and exert long-range effects. Here, we will discuss classical and emerging technologies by which one can understand the role of binding of specific transcription factors in regulation of transcription of FOXO genes. DOI: 10.1007/978-1-4939-8900-3_3 PMID: 30414142 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35020192
1. Muscle Nerve. 2022 Apr;65(4):415-421. doi: 10.1002/mus.27493. Epub 2022 Jan 25. Systemic manifestations and symptom burden of facioscapulohumeral muscular dystrophy in a referral cohort. Kelly CR(1), Saw JL(1), Thapa P(2), Mandrekar J(2), Naddaf E(1). Author information: (1)Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA. (2)Department of Internal Medicine-Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA. INTRODUCTION/AIMS: The full spectrum of the clinical phenotype of facioscapulohumeral muscular dystrophy (FSHD), beyond skeletal muscle weakness, remains poorly characterized. In this study, we describe systemic manifestations and symptom burden in a large series of FSHD patients. METHODS: We performed a retrospective chart review of FSHD patients seen at our institution between 2000 and 2017. We reviewed patients' responses to a comprehensive review of symptoms and the results of diagnostic testing for sensorineural hearing loss, cardiac disease, dysphagia, ocular abnormalities, and respiratory insufficiency. We assessed the association between disease manifestations and age of onset, genetic profile, and disease duration. RESULTS: We identified 87 patients with FSHD. The most common reported symptoms included pain (71%), difficulty sleeping (41%), headaches (27%), and altered mood (24%). When tested, 7 of 16 (44%) patients had sensorineural hearing loss, 20 of 60 (33%) had cardiac arrhythmias or conduction defects, 17 of 45 (38%) had echocardiogram abnormalities, 12 of 25 (48%) had reduced forced vital capacity, and 4 of 10 (40%) had oropharyngeal dysphagia. However, patients with these abnormalities represented 8%, 23%, 20%, 14%, and 5% of total number of patients, respectively, as uniform screening was lacking. Ocular pathology attributable to FSHD was not detected. DISCUSSION: FSHD demonstrates a broad clinical phenotype. Increased vigilance among neurologists to screen for systemic manifestations of the disease is warranted. More uniform screening and future population-based studies are needed to compare findings in FSHD patients with the general population. © 2022 Wiley Periodicals LLC. DOI: 10.1002/mus.27493 PMID: 35020192 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/15050443
1. J Neurol Sci. 2004 Apr 15;219(1-2):89-93. doi: 10.1016/j.jns.2003.12.010. FSHD-like patients without 4q35 deletion. Yamanaka G(1), Goto K, Ishihara T, Oya Y, Miyajima T, Hoshika A, Nishino I, Hayashi YK. Author information: (1)Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8502, Japan. Facioscapulohumeral muscular dystrophy (FSHD) is characterized by progressive weakness and wasting of facial, shoulder-girdle and upper arm muscles. Despite of the characteristic clinical features, the diagnosis of FSHD is sometimes difficult because clinical symptoms are extremely variable including facial sparing type, limb-girdle type, and distal myopathy type. Most of the FSHD patients have a deletion in the subtelomeric region of chromosome 4q35 (FSHMD1A), however the linkage analysis in some families suggested genetic heterogeneity. In the present study, we identified 40 patients without a deletion in the 4q35 region (non-4q35del) among 200 Japanese patients who were clinically suspected to have FHSD. All non-4q35del patients had shoulder-girdle weakness and 75% also had facial weakness. Eight patients showed clinical features that were indistinguishable from FSHD, but two of them had Becker muscular dystrophy. FSHD is clinically, and most likely genetically, as well, variable. Other forms of muscular dystrophy can also mimic FSHD. DOI: 10.1016/j.jns.2003.12.010 PMID: 15050443 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/22907234
1. Muscle Nerve. 2012 Sep;46(3):421-5. doi: 10.1002/mus.23358. Focal and other unusual presentations of facioscapulohumeral muscular dystrophy. Hassan A(1), Jones LK Jr, Milone M, Kumar N. Author information: (1)Department of Neurology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, Minnesota 55905, USA. INTRODUCTION: Facioscapulohumeral dystrophy (FSHD) presents classically with facial and shoulder-girdle weakness. We report focal atypical presentations of FSHD. Our aim was to identify focal/unusual phenotypes in genetically confirmed FSHD cases. METHODS: We undertook a retrospective review of an academic center database of the period from 1996 to 2011. Of 139 FSHD cases, 7 had atypical genetically confirmed disease. Clinical data were abstracted. RESULTS: Seven cases (4 men) had a mean age of 37 years at onset (range 18-63 years) and mean 43 years at diagnosis (range 20-74 years). Presenting symptoms were monomelic lower limb (n = 3) or upper limb (n = 2) atrophy, or axial weakness (n = 2). Five patients had focal weakness on examination. CK was normal to borderline high. Two patients had a relative with FSHD. Coexistent unusual features included dyspnea (n = 1), S1 radicular pain with calf atrophy (n = 2), and peripheral neuropathy (n = 1). Almost all patients had myopathic EMG changes. DNA analysis showed a D4Z4 EcoRI fragment size ranging from 20 to 37 kilobases. CONCLUSIONS: FSHD may present with focal weakness, dyspnea and myopathic EMG changes. These findings should raise the possibility of FSHD. Copyright © 2012 Wiley Periodicals, Inc. DOI: 10.1002/mus.23358 PMID: 22907234 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34675094
1. Neurology. 2021 Nov 23;97(21):e2103-e2113. doi: 10.1212/WNL.0000000000012882. Epub 2021 Oct 21. Natural History of Facioscapulohumeral Dystrophy in Children: A 2-Year Follow-up. Dijkstra JN(1), Goselink RJM(1), van Alfen N(1), de Groot IJM(1), Pelsma M(1), van der Stoep N(1), Theelen T(1), van Engelen BGM(1), Voermans NC(1), Erasmus CE(2). Author information: (1)From the Departments of Neurology (J.N.D., N.v.A., B.G.M.v.E., N.C.V.) and Rehabilitation (I.J.M.d.G., M.P.), Donders Centre of Neuroscience, Department of Pediatric Neurology (J.N.D., C.E.E.), Amalia Children's Hospital, and Department of Ophthalmology (T.T.), Radboud University Medical Centre, Nijmegen, the Netherlands; Department of Neurology (R.J.M.G.), Jönköping, Region Jönköping County, and Department of Biomedical and Clinical Sciences (R.J.M.G.), Linköping University, Linköping, Sweden; and Department of Clinical Genetics (N.v.d.S.), Leiden University Medical Centre, the Netherlands. (2)From the Departments of Neurology (J.N.D., N.v.A., B.G.M.v.E., N.C.V.) and Rehabilitation (I.J.M.d.G., M.P.), Donders Centre of Neuroscience, Department of Pediatric Neurology (J.N.D., C.E.E.), Amalia Children's Hospital, and Department of Ophthalmology (T.T.), Radboud University Medical Centre, Nijmegen, the Netherlands; Department of Neurology (R.J.M.G.), Jönköping, Region Jönköping County, and Department of Biomedical and Clinical Sciences (R.J.M.G.), Linköping University, Linköping, Sweden; and Department of Clinical Genetics (N.v.d.S.), Leiden University Medical Centre, the Netherlands corrie.erasmus@radboudumc.nl. BACKGROUND AND OBJECTIVES: Data on the natural history of facioscapulohumeral dystrophy (FSHD) in childhood are limited and critical for improved patient care and clinical trial readiness. Our objective was to describe the disease course of FSHD in children. METHODS: We performed a nationwide, single-center, prospective cohort study of FSHD in childhood assessing muscle functioning, imaging, and quality of life over 2 years of follow-up. RESULTS: We included 20 children with genetically confirmed FSHD who were 2 to 17 years of age. Overall, symptoms were slowly progressive, and the mean FSHD clinical score increased from 2.1 to 2.8 (p = 0.003). The rate of progression was highly variable. At baseline, 16 of 20 symptomatic children had facial weakness; after 2 years, facial weakness was observed in 19 of 20 children. Muscle strength did not change between baseline and follow-up. The most frequently and most severely affected muscles were the trapezius and deltoid. The functional exercise capacity, measured with the 6-minute walk test, improved. Systemic features were infrequent and nonprogressive. Weakness-associated complications such as lumbar hyperlordosis and dysarthria were common, and their prevalence increased during follow-up. Pain and fatigue were frequent complaints in children, and their prevalence also increased during follow-up. Muscle ultrasonography revealed a progressive increase in echogenicity. DISCUSSION: FSHD in childhood has a slowly progressive but variable course over 2 years of follow-up. The most promising outcome measures to detect progression were the FSHD clinical score and muscle ultrasonography. Despite this disease progression, an improvement on functional capacity may still occur as the child grows up. Pain, fatigue, and a decreased quality of life were common symptoms and need to be addressed in the management of childhood FSHD. Our data can be used to counsel patients and as baseline measures for treatment trials in childhood FSHD. Copyright © 2021 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology. DOI: 10.1212/WNL.0000000000012882 PMCID: PMC8610619 PMID: 34675094 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/32008231
1. Ther Innov Regul Sci. 2020 Jan;54(1):144-150. doi: 10.1007/s43441-019-00038-w. Epub 2020 Jan 6. Updating the Clinical Picture of Facioscapulohumeral Muscular Dystrophy: Ramifications for Drug Development With Potential Solutions. Huml RA(1), Uspenskaya-Cadoz O(2), Dawson J(3), Slifer Z(4). Author information: (1)IQVIA, Durham, North Carolina, 27703, USA. Raymond.huml@iqvia.com. (2)IQVIA, Durham, North Carolina, 27703, USA. (3)Consultant, Durham, NC, USA. (4)North Carolina State University's College of Veterinary Medicine, Raleigh, North Carolina, USA. BACKGROUND: Facioscapulohumeral muscular dystrophy (FSHD) is a complex, inheritable, and rare muscle disease that affects the entire body. The major symptom of FSHD is progressive weakening and loss of skeletal muscles. The usual location of these weaknesses at onset is the origin of the name: face (facio), shoulder girdle (scapulo), and upper arms (humeral). FSHD appears to have varying molecular and genetic determinants with commensurate differences in disease progression. METHODS: Facioscapulohumeral muscular dystrophy (MD) is probably the most prevalent form of MD but has neither disease-modifying treatments nor a cure. As the mechanism of action becomes further elucidated, more biopharmaceutical companies are investing capital into finding treatments for patients with FSHD. Sponsors of treatments for FSHD patients should be aware of some of the common misconceptions associated with FSHD drug development with the goal of optimizing the chance to prove safety and efficacy for each potential treatment for FSHD in the clinical trial setting. RESULTS: Four major topics with potential clinical manifestations for patients with FSHD will be discussed related to muscle weakness, respiratory issues, animal models and prevalence. CONCLUSION: The authors offer multiple solutions to help counteract misconceptions with each scenario during clinical trial drug development. DOI: 10.1007/s43441-019-00038-w PMID: 32008231 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/31409737
1. Neurology. 2019 Sep 17;93(12):e1180-e1192. doi: 10.1212/WNL.0000000000008123. Epub 2019 Aug 13. Patient-Reported Symptoms in Facioscapulohumeral Muscular Dystrophy (PRISM-FSHD). Hamel J(1), Johnson N(2), Tawil R(2), Martens WB(2), Dilek N(2), McDermott MP(2), Heatwole C(2). Author information: (1)From the Department of Neurology (J.H., R.T., W.B.M., N.D., M.P.M., C.H.) and Department of Biostatistics and Computational Biology (M.P.M.), University of Rochester Medical Center, NY; and Department of Neurology (N.J.), Virginia Commonwealth University, Richmond. johanna_hamel@urmc.rochester.edu. (2)From the Department of Neurology (J.H., R.T., W.B.M., N.D., M.P.M., C.H.) and Department of Biostatistics and Computational Biology (M.P.M.), University of Rochester Medical Center, NY; and Department of Neurology (N.J.), Virginia Commonwealth University, Richmond. OBJECTIVE: To determine the frequency and relative importance of the most meaningful symptoms in facioscapulohumeral muscular dystrophy (FSHD) and to identify the demographic and clinical features that are associated with the greatest disease burden in this population. METHODS: We performed a cross-sectional study involving 328 participants with FSHD. Collectively, participants reported the prevalence and relative importance of 274 symptoms and 15 symptomatic themes. We assessed the association between symptomatic theme prevalence and participants' age, sex, disease duration, pain level, employment status, and education. RESULTS: Participants answered >48,000 questions regarding their disease burden. The symptomatic themes with the highest prevalence in our sample were problems with shoulders or arms (96.9%), limitations with activities (94.7%), core weakness (93.8%), fatigue (93.8%), limitations with mobility and walking (93.6%), changed body image due to the disease (91.6%), and pain (87.7%). Problems with shoulders and arms and limitations with mobility and walking had the greatest effect on participants' lives. Employment status and the report of pain had the most extensive association with the prevalence of symptoms, with employment being associated with 8 of 15 of the symptomatic themes and pain being associated with 7 of 15 of the symptomatic themes. Men and women with FSHD experienced a similar prevalence of all symptomatic themes. CONCLUSIONS: Adults with FSHD experience a variety of symptoms that play an important role in their disease burden. These symptoms have a variable prevalence and importance in the FSHD population and are associated with disease duration, employment status, and pain level. © 2019 American Academy of Neurology. DOI: 10.1212/WNL.0000000000008123 PMCID: PMC6808532 PMID: 31409737 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/27530735
1. BMC Neurol. 2016 Aug 17;16:138. doi: 10.1186/s12883-016-0664-6. Facioscapulohumeral dystrophy in children: design of a prospective, observational study on natural history, predictors and clinical impact (iFocus FSHD). Goselink RJ(1), Schreuder TH(2), Mul K(2), Voermans NC(2), Pelsma M(3), de Groot IJ(3), van Alfen N(2), Franck B(4), Theelen T(5), Lemmers RJ(6), Mah JK(7), van der Maarel SM(6), van Engelen BG(2), Erasmus CE(2). Author information: (1)Department of Neurology, Donders Center for Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands. Rianne.Goselink@radboudumc.nl. (2)Department of Neurology, Donders Center for Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands. (3)Department of Rehabilitation, Donders Center for Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands. (4)Department of Clinical audiology, Radboud University Medical Center, Nijmegen, The Netherlands. (5)Department of Op Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands. (6)Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands. (7)Department of Paediatric Neurology, Alberta Children's Hospital, Calgary, Canada. BACKGROUND: Facioscapulohumeral muscular dystrophy (FSHD; OMIM 158900 & 158901) is a progressive skeletal muscle dystrophy, characterized by an autosomal dominant inheritance pattern. One of the major unsolved questions in FSHD is the marked clinical heterogeneity, ranging from asymptomatic individuals to severely affected patients with an early onset. An estimated 10% of FSHD patients have an early onset (onset before 10 years of age) and are traditionally classified as infantile FSHD. This subgroup is regarded as severely affected and extra-muscular symptoms, such as hearing loss and retinopathy, are frequently described. However, information on the prevalence, natural history and clinical management of early onset FSHD is currently lacking, thereby hampering adequate patient counselling and management. Therefore, a population-based prospective cohort study on FSHD in children is highly needed. METHODS/DESIGN: This explorative study aims to recruit all children (aged 0-17 years) with a genetically confirmed diagnosis of FSHD in The Netherlands. The children will be assessed at baseline and at 2-year follow-up. The general aim of the study is the description of the clinical features and genetic characteristics of this paediatric cohort. The primary outcome is the motor function as measured by the Motor Function Measure. Secondary outcomes include quantitative and qualitative description of the clinical phenotype, muscle imaging, genotyping and prevalence estimations. The ultimate objective will be a thorough description of the natural history, predictors of disease severity and quality of life in children with FSHD. DISCUSSION: The results of this population-based study are vital for adequate patient management and clinical trial-readiness. Furthermore, this study is expected to provide additional insight in the epigenetic and environmental disease modifying factors. In addition to improve counselling, this could contribute to unravelling the aetiology of FSHD. TRIAL REGISTRATION: clinicaltrials.gov NCT02625662. DOI: 10.1186/s12883-016-0664-6 PMCID: PMC4988042 PMID: 27530735 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/22796148
1. Free Radic Biol Med. 2012 Sep 1;53(5):1068-79. doi: 10.1016/j.freeradbiomed.2012.06.041. Epub 2012 Jul 11. Functional muscle impairment in facioscapulohumeral muscular dystrophy is correlated with oxidative stress and mitochondrial dysfunction. Turki A(1), Hayot M, Carnac G, Pillard F, Passerieux E, Bommart S, Raynaud de Mauverger E, Hugon G, Pincemail J, Pietri S, Lambert K, Belayew A, Vassetzky Y, Juntas Morales R, Mercier J, Laoudj-Chenivesse D. Author information: (1)Université Montpellier 1 et Université Montpellier 2, INSERM, U1046, Montpellier, F-34000, France. Facioscapulohumeral muscular dystrophy (FSHD), the most frequent muscular dystrophy, is an autosomal dominant disease. In most individuals with FSHD, symptoms are restricted to muscles of the face, arms, legs, and trunk. FSHD is genetically linked to contractions of the D4Z4 repeat array causing activation of several genes. One of these maps in the repeat itself and expresses the DUX4 (the double homeobox 4) transcription factor causing a gene deregulation cascade. In addition, analyses of the RNA or protein expression profiles in muscle have indicated deregulations in the oxidative stress response. Since oxidative stress affects peripheral muscle function, we investigated mitochondrial function and oxidative stress in skeletal muscle biopsies and blood samples from patients with FSHD and age-matched healthy controls, and evaluated their association with physical performances. We show that specifically, oxidative stress (lipid peroxidation and protein carbonylation), oxidative damage (lipofuscin accumulation), and antioxidant enzymes (catalase, copper-zinc-dependent superoxide dismutase, and glutathione reductase) were higher in FSHD than in control muscles. FSHD muscles also presented abnormal mitochondrial function (decreased cytochrome c oxidase activity and reduced ATP synthesis). In addition, the ratio between reduced (GSH) and oxidized glutathione (GSSG) was strongly decreased in all FSHD blood samples as a consequence of GSSG accumulation. Patients with FSHD also had reduced systemic antioxidative response molecules, such as low levels of zinc (a SOD cofactor), selenium (a GPx cofactor involved in the elimination of lipid peroxides), and vitamin C. Half of them had a low ratio of gamma/alpha tocopherol and higher ferritin concentrations. Both systemic oxidative stress and mitochondrial dysfunction were correlated with functional muscle impairment. Mitochondrial ATP production was significantly correlated with both quadriceps endurance (T(LimQ)) and maximal voluntary contraction (MVC(Q)) values (rho=0.79, P=0.003; rho=0.62, P=0.05, respectively). The plasma concentration of oxidized glutathione was negatively correlated with the T(LimQ), MVC(Q) values, and the 2-min walk distance (MWT) values (rho=-0.60, P=0.03; rho=-0.56, P=0.04; rho=-0.93, P<0.0001, respectively). Our data characterized oxidative stress in patients with FSHD and demonstrated a correlation with their peripheral skeletal muscle dysfunction. They suggest that antioxidants that might modulate or delay oxidative insult may be useful in maintaining FSHD muscle functions. Copyright © 2012 Elsevier Inc. All rights reserved. DOI: 10.1016/j.freeradbiomed.2012.06.041 PMID: 22796148 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/10864616
1. J Neurol Neurosurg Psychiatry. 2000 Jul;69(1):114-6. doi: 10.1136/jnnp.69.1.114. Extension of the clinical range of facioscapulohumeral dystrophy: report of six cases. van der Kooi AJ(1), Visser MC, Rosenberg N, van den Berg-Vos R, Wokke JH, Bakker E, de Visser M. Author information: (1)Department of Neurology, Academic Medical Center, University of Amsterdam, The Netherlands. a.j.kooi@amc.uva.nl Consensual diagnostic criteria for facioscapulohumeral dystrophy (FSHD) include onset of the disease in facial or shoulder girdle muscles, facial weakness in more than 50% of affected family members, autosomal dominant inheritance in familial cases, and evidence of myopathic disease in at least one affected member without biopsy features specific to alternative diagnoses. Six patients did not meet most of these criteria but were diagnosed as FSHD by DNA testing, which showed small EcoRI fragments on chromosome 4q. Their clinical signs and symptoms and results of auxiliary investigations are reported. The patients presented with foot extensor, thigh, or calf muscle weakness. None of them had apparent facial weakness, only one complained of weakness in the shoulders, none had a positive family history. Expert physical examination, however, showed a typical facial expression, an abnormal shoulder configuration on lifting the arms, or scapular winging. This raised the suspicion of FSHD, whereupon DNA analysis was done. In conclusion, the clinical expression of FSHD is much broader than indicated by the nomenclature. The possibility to perform DNA tests is likely to greatly expand the clinical range of FSHD. DOI: 10.1136/jnnp.69.1.114 PMCID: PMC1737027 PMID: 10864616 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/33096728
1. Int J Mol Sci. 2020 Oct 21;21(20):7783. doi: 10.3390/ijms21207783. Early-Onset Infantile Facioscapulohumeral Muscular Dystrophy: A Timely Review. Chen TH(1)(2)(3), Wu YZ(2), Tseng YH(2). Author information: (1)Section of Neurobiology, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA. (2)Department of Pediatrics, Division of Pediatric Emergency, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan. (3)School of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan. Facioscapulohumeral muscular dystrophy (FSHD)-the worldwide third most common inherited muscular dystrophy caused by the heterozygous contraction of a 3.3 kb tandem repeat (D4Z4) on a chromosome with a 4q35 haplotype-is a progressive genetic myopathy with variable onset of symptoms, distribution of muscle weakness, and clinical severity. While much is known about the clinical course of adult FSHD, data on the early-onset infantile phenotype, especially on the progression of the disease, are relatively scarce. Contrary to the classical form, patients with infantile FSHD more often have a rapid decline in muscle wasting and systemic features with multiple extramuscular involvements. A rough correlation between the phenotypic severity of FSHD and the D4Z4 repeat size has been reported, and the majority of patients with infantile FSHD obtain a very short D4Z4 repeat length (one to three copies, EcoRI size 10-14 kb), in contrast to the classical, slowly progressive, form of FSHD (15-38 kb). With the increasing identifications of case reports and the advance in genetic diagnostics, recent studies have suggested that the infantile variant of FSHD is not a genetically separate entity but a part of the FSHD spectrum. Nevertheless, many questions about the clinical phenotype and natural history of infantile FSHD remain unanswered, limiting evidence-based clinical management. In this review, we summarize the updated research to gain insight into the clinical spectrum of infantile FSHD and raise views to improve recognition and understanding of its underlying pathomechanism, and further, to advance novel treatments and standard care methods. DOI: 10.3390/ijms21207783 PMCID: PMC7589635 PMID: 33096728 [Indexed for MEDLINE] Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/23785297
1. PLoS Genet. 2013 Jun;9(6):e1003550. doi: 10.1371/journal.pgen.1003550. Epub 2013 Jun 13. Deregulation of the protocadherin gene FAT1 alters muscle shapes: implications for the pathogenesis of facioscapulohumeral dystrophy. Caruso N(1), Herberth B, Bartoli M, Puppo F, Dumonceaux J, Zimmermann A, Denadai S, Lebossé M, Roche S, Geng L, Magdinier F, Attarian S, Bernard R, Maina F, Levy N, Helmbacher F. Author information: (1)Aix-Marseille Université, CNRS, IBDML UMR 7288, Parc Scientifique de Luminy, Case 907, Marseille, France. Generation of skeletal muscles with forms adapted to their function is essential for normal movement. Muscle shape is patterned by the coordinated polarity of collectively migrating myoblasts. Constitutive inactivation of the protocadherin gene Fat1 uncoupled individual myoblast polarity within chains, altering the shape of selective groups of muscles in the shoulder and face. These shape abnormalities were followed by early onset regionalised muscle defects in adult Fat1-deficient mice. Tissue-specific ablation of Fat1 driven by Pax3-cre reproduced muscle shape defects in limb but not face muscles, indicating a cell-autonomous contribution of Fat1 in migrating muscle precursors. Strikingly, the topography of muscle abnormalities caused by Fat1 loss-of-function resembles that of human patients with facioscapulohumeral dystrophy (FSHD). FAT1 lies near the critical locus involved in causing FSHD, and Fat1 mutant mice also show retinal vasculopathy, mimicking another symptom of FSHD, and showed abnormal inner ear patterning, predictive of deafness, reminiscent of another burden of FSHD. Muscle-specific reduction of FAT1 expression and promoter silencing was observed in foetal FSHD1 cases. CGH array-based studies identified deletion polymorphisms within a putative regulatory enhancer of FAT1, predictive of tissue-specific depletion of FAT1 expression, which preferentially segregate with FSHD. Our study identifies FAT1 as a critical determinant of muscle form, misregulation of which associates with FSHD. DOI: 10.1371/journal.pgen.1003550 PMCID: PMC3681729 PMID: 23785297 [Indexed for MEDLINE] Conflict of interest statement: The authors have declared that no competing interests exist.
http://www.ncbi.nlm.nih.gov/pubmed/34964760
1. Medicine (Baltimore). 2021 Nov 24;100(47):e27907. doi: 10.1097/MD.0000000000027907. A pediatric case report and literature review of facioscapulohumeral muscular dystrophy type1. Xiao T(1), Yang H(2), Gan S(2), Wu L(2). Author information: (1)Department of Pediatric Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China. (2)Department of Pediatric Neurology, Hunan Children's Hospital, Changsha, Hunan, PR China. RATIONALE: Early-onset facioscapulohumeral muscular dystrophy (FSHD) is defined as facial weakness before the age of 5 and shoulder weakness before the age of 10. Early-onset facioscapulohumeral muscular dystrophy is relatively rare in the clinic. This onset is relatively early, the symptoms are serious, and it is likely to be accompanied by retinal vascular disease, sensorineural deafness, epilepsy and other extramuscular multisystem diseases. We report the clinical characteristics of 2 patients with early-onset facial and shoulder brachial muscular dystrophy to improve clinicians' understanding of this particular condition. PATIENT CONCERNS: We report 2 pediatric patients with FSHD type 1. Patient 1 is an 11-year-old boy with reduced facial expression for 9 years and proximal muscle weakness for 6 years. Patient 2 is a 4-year and 6-month-old girl with developmental delay for 3 years and facial weakness for 1 year. DIAGNOSIS: According to the clinical manifestations and molecular genetic testing (such as Southern blot analysis), the patients were diagnosed with early-onset FSHD1. INTERVENTIONS: The patients received cocktail therapy (vitamin B1 tablets, vitamin B2 tablets, vitamin B6 tablets, vitamin C tablets, vitamin E tablets, idebenone tablets, etc.) to improve their muscle metabolism. OUTCOMES: Both patients' condition did not improve after being given cocktail treatment. According to a recent follow-up, the symptoms of facial weakness and proximal muscle weakness were aggravated. LESSONS: Early-onset FSHD presents early and has frequent systemic features, and it is a severe subtype of FSHD. Early identification and genetic diagnosis should be performed to improve patient prognosis. Copyright © 2021 the Author(s). Published by Wolters Kluwer Health, Inc. DOI: 10.1097/MD.0000000000027907 PMCID: PMC8615324 PMID: 34964760 [Indexed for MEDLINE] Conflict of interest statement: The authors have no conflicts of interests to disclose.
http://www.ncbi.nlm.nih.gov/pubmed/11353419
1. Muscle Nerve. 2001 Mar;24(3):352-6. doi: 10.1002/1097-4598(200103)24:3<352::aid-mus1005>3.0.co;2-m. Unusual clinical presentations in patients harboring the facioscapulohumeral dystrophy 4q35 deletion. Felice KJ(1), Moore SA. Author information: (1)Department of Neurology, University of Connecticut School of Medicine, Farmington, Connecticut 06035-1840, USA. felice@nso.uchc.edu Facioscapulohumeral dystrophy (FSHD) is a dominantly inherited myopathy usually associated with a deletion at locus 4q35. Typically, FSHD patients present with a recognizable constellation of signs including weakness of facial, shoulder and pelvic girdle, humeral, and anterior foreleg muscles; preservation of some muscles including the deltoids; and other characteristic features including prominent scapular winging, anterior axillary folds, and horizontally positioned clavicles. We performed clinical and FSHD genetic studies on four patients with atypical clinical features who were cared for at a regional neuromuscular center. The four patients, each harboring 4q35 deletions, presented with atypical phenotypes including facial-sparing scapular myopathy, limb-girdle muscular dystrophy, distal myopathy, and asymmetric brachial weakness. This report demonstrates the expanding clinical heterogeneity in patients harboring the 4q35 deletion. Copyright 2001 John Wiley & Sons, Inc. DOI: 10.1002/1097-4598(200103)24:3<352::aid-mus1005>3.0.co;2-m PMID: 11353419 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/32278354
1. Skelet Muscle. 2020 Apr 11;10(1):8. doi: 10.1186/s13395-020-00227-4. Transgenic mice expressing tunable levels of DUX4 develop characteristic facioscapulohumeral muscular dystrophy-like pathophysiology ranging in severity. Jones TI(1), Chew GL(2)(3)(4), Barraza-Flores P(1), Schreier S(1), Ramirez M(1), Wuebbles RD(1), Burkin DJ(1), Bradley RK(2)(3), Jones PL(5). Author information: (1)Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, NV, 89557, USA. (2)Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA. (3)Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA. (4)Current Address: The Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore. (5)Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, NV, 89557, USA. peterjones@med.unr.edu. BACKGROUND: All types of facioscapulohumeral muscular dystrophy (FSHD) are caused by the aberrant activation of the somatically silent DUX4 gene, the expression of which initiates a cascade of cellular events ultimately leading to FSHD pathophysiology. Typically, progressive skeletal muscle weakness becomes noticeable in the second or third decade of life, yet there are many individuals who are genetically FSHD but develop symptoms much later in life or remain relatively asymptomatic throughout their lives. Conversely, FSHD may clinically present prior to 5-10 years of age, ultimately manifesting as a severe early-onset form of the disease. These phenotypic differences are thought to be due to the timing and levels of DUX4 misexpression. METHODS: FSHD is a dominant gain-of-function disease that is amenable to modeling by DUX4 overexpression. We have recently created a line of conditional DUX4 transgenic mice, FLExDUX4, that develop a myopathy upon induction of human DUX4-fl expression in skeletal muscle. Here, we use the FLExDUX4 mouse crossed with the skeletal muscle-specific and tamoxifen-inducible line ACTA1-MerCreMer to generate a highly versatile bi-transgenic mouse model with chronic, low-level DUX4-fl expression and cumulative mild FSHD-like pathology that can be reproducibly induced to develop more severe pathology via tamoxifen induction of DUX4-fl in skeletal muscles. RESULTS: We identified conditions to generate FSHD-like models exhibiting reproducibly mild, moderate, or severe DUX4-dependent pathophysiology and characterized progression of pathology. We assayed DUX4-fl mRNA and protein levels, fitness, strength, global gene expression, and histopathology, all of which are consistent with an FSHD-like myopathic phenotype. Importantly, we identified sex-specific and muscle-specific differences that should be considered when using these models for preclinical studies. CONCLUSIONS: The ACTA1-MCM;FLExDUX4 bi-transgenic mouse model has mild FSHD-like pathology and detectable muscle weakness. The onset and progression of more severe DUX4-dependent pathologies can be controlled via tamoxifen injection to increase the levels of mosaic DUX4-fl expression, providing consistent and readily screenable phenotypes for assessing therapies targeting DUX4-fl mRNA and/or protein and are useful to investigate certain conserved downstream FSHD-like pathophysiology. Overall, this model supports that DUX4 expression levels in skeletal muscle directly correlate with FSHD-like pathology by numerous metrics. DOI: 10.1186/s13395-020-00227-4 PMCID: PMC7149937 PMID: 32278354 [Indexed for MEDLINE] Conflict of interest statement: The authors declare that they have no competing interests.
http://www.ncbi.nlm.nih.gov/pubmed/31025273
1. Sleep Breath. 2019 Sep;23(3):899-906. doi: 10.1007/s11325-019-01843-1. Epub 2019 Apr 26. Sleep-related breathing disorders in facioscapulohumeral dystrophy. Runte M(#)(1), Spiesshoefer J(#)(1), Heidbreder A(1), Dreher M(2), Young P(3), Brix T(4), Boentert M(5). Author information: (1)Department of Neurology, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149, Muenster, Germany. (2)Department of Pneumology and Intensive Care Medicine, University Hospital RWTH Aachen, Aachen, Germany. (3)Medical Park Klinik Reithofpark, Neurology, Bad Feilnbach, Germany. (4)Institute of Medical Informatics, University of Muenster, Muenster, Germany. (5)Department of Neurology, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149, Muenster, Germany. matthias.boentert@ukmuenster.de. (#)Contributed equally Comment in Sleep Breath. 2020 Jun;24(2):603-604. doi: 10.1007/s11325-019-01991-4. Sleep Breath. 2020 Jun;24(2):675-676. doi: 10.1007/s11325-019-01992-3. PURPOSE: Severe manifestations of facioscapulohumeral dystrophy (FSHD) may be associated with sleep-disordered breathing (SDB), including obstructive sleep apnea (OSA) and nocturnal hypoventilation (NH), but prevalence data are scarce. In patients with respiratory muscle weakness, detection of NH can be facilitated by transcutaneous capnometry, but respective data derived from FSHD patients have not yet been published. METHODS: We collected sleep studies and capnometry recordings from 31 adult patients with genetically confirmed FSHD who were admitted to our sleep laboratory for first-ever evaluation of sleep-related breathing. Indications for admission included non-restorative sleep, morning headache, or excessive daytime sleepiness. In addition, sleep studies were initiated if symptoms or signs of respiratory muscle weakness were present. Thirty-one subjects with insomnia served as controls for comparison of respiratory measures during sleep. RESULTS: In the FSHD group, 17/31 (55%) patients showed OSA and 8 (26%) had NH. NH would have been missed in 7/8 patients if only oximetry criteria of hypoventilation had been applied. Capnography results were correlated with disease severity as reflected by the Clinical Severity Score (CSS). Non-invasive ventilation (NIV) was started in 6 patients with NH and 3 individuals with OSA. Nocturnal continuous positive airway pressure was administered to 2 patients, and positional therapy was sufficient in 4 individuals. In patients initiated on NIV, nocturnal gas exchange already improved in the first night of treatment. CONCLUSIONS: SDB is common in adult patients with FSHD complaining of sleep-related symptoms. It may comprise OSA, NH, and most often, the combination of both. Sleep-related hypercapnia is associated with disease severity. Transcutaneous capnometry is superior to pulse oximetry for detection of NH. DOI: 10.1007/s11325-019-01843-1 PMID: 31025273 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34559225
1. Hum Mol Genet. 2022 Mar 3;31(5):748-760. doi: 10.1093/hmg/ddab250. High-resolution breakpoint junction mapping of proximally extended D4Z4 deletions in FSHD1 reveals evidence for a founder effect. Lemmers RJLF(1), van der Vliet PJ(1), Granado DSL(1), van der Stoep N(2), Buermans H(1), van Schendel R(1), Schimmel J(1), de Visser M(3), van Coster R(4), Jeanpierre M(5), Laforet P(6), Upadhyaya M(7), van Engelen B(8), Sacconi S(9), Tawil R(10), Voermans NC(8), Rogers M(7), van der Maarel SM(1). Author information: (1)Department of Human Genetics, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands. (2)Department of Clinical Genetics, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands. (3)Department of Neurology, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands. (4)Department of Pediatrics Neurology, Ghent University Hospital, Ghent, 9000, Belgium. (5)APHP-Hôpitaux de Paris, Université de Paris, Paris, 75400, France. (6)Neurology Department, Raymond-Poincaré Hospital, Garches, 92380, France. (7)Department of Medical Genetics, Cardiff University, Cardiff, CF10 3AT, UK. (8)Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, 6500 HB, The Netherlands. (9)Centre de référence des Maladies neuromusculaires, Nice University Hospital, Nice, 06000, France. (10)Department of Neurology, University of Rochester Medical Center, Rochester, NY, NY 14642, USA. Facioscapulohumeral muscular dystrophy (FSHD) is an inherited myopathy clinically characterized by weakness in the facial, shoulder girdle and upper a muscles. FSHD is caused by chromatin relaxation of the D4Z4 macrosatellite repeat, mostly by a repeat contraction, facilitating ectopic expression of DUX4 in skeletal muscle. Genetic diagnosis for FSHD is generally based on the sizing and haplotyping of the D4Z4 repeat on chromosome 4 by Southern blotting (SB), molecular combing or single-molecule optical mapping, which is usually straight forward but can be complicated by atypical rearrangements of the D4Z4 repeat. One of these rearrangements is a D4Z4 proximally extended deletion (DPED) allele, where not only the D4Z4 repeat is partially deleted, but also sequences immediately proximal to the repeat are lost, which can impede accurate diagnosis in all genetic methods. Previously, we identified several DPED alleles in FSHD and estimated the size of the proximal deletions by a complex pulsed-field gel electrophoresis and SB strategy. Here, using the next-generation sequencing, we have defined the breakpoint junctions of these DPED alleles at the base pair resolution in 12 FSHD families and 4 control individuals facilitating a PCR-based diagnosis of these DPED alleles. Our resultsshow that half of the DPED alleles are derivates of an ancient founder allele. For some DPED alleles, we found that genetic elements are deleted such as DUX4c, FRG2, DBE-T and myogenic enhancers necessitating re-evaluation of their role in FSHD pathogenesis. © The Author(s) 2021. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com. DOI: 10.1093/hmg/ddab250 PMCID: PMC8895739 PMID: 34559225 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/36030628
1. Redox Biol. 2022 Oct;56:102450. doi: 10.1016/j.redox.2022.102450. Epub 2022 Aug 22. ANT1 overexpression models: Some similarities with facioscapulohumeral muscular dystrophy. Arbogast S(1), Kotzur H(2), Frank C(2), Compagnone N(3), Sutra T(4), Pillard F(5), Pietri S(6), Hmada N(1), Moussa DMA(7), Bride J(1), Françonnet S(1), Mercier J(4), Cristol JP(4), Dabauvalle MC(2), Laoudj-Chenivesse D(8). Author information: (1)PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France. (2)Imaging Core Facility, Biocenter, University of Würzburg, Am Hubland, 97074, Würzburg, Germany. (3)Innovative Concepts in Drug Development (ICDD), Gemenos, France. (4)PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France; CHRU de Montpellier, Montpellier, France. (5)Sport Medicine Department, University Sport Clinic, Pierre Paul Riquet University Hospital, Toulouse, France; Institute of Metabolic and Cardiovascular Diseases, Joint Research Unit 1048 INSERM Adipolab Unit - Paul Sabatier University, Toulouse, France. (6)Aix Marseille Univ, CNRS, ICR, UMR, 7273, Marseille, France. (7)IRMB, Univ Montpellier, INSERM (METAMONTP), Montpellier, France. (8)PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France; CHRU de Montpellier, Montpellier, France. Electronic address: dalila.laoudj-chenivesse@inserm.fr. Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant disorder characterized by progressive muscle weakness. Adenine nucleotide translocator 1 (ANT1), the only 4q35 gene involved in mitochondrial function, is strongly expressed in FSHD skeletal muscle biopsies. However, its role in FSHD is unclear. In this study, we evaluated ANT1 overexpression effects in primary myoblasts from healthy controls and during Xenopus laevis organogenesis. We also compared ANT1 overexpression effects with the phenotype of FSHD muscle cells and biopsies. Here, we report that the ANT1 overexpression-induced phenotype presents some similarities with FSHD muscle cells and biopsies. ANT1-overexpressing muscle cells showed disorganized morphology, altered cytoskeletal arrangement, enhanced mitochondrial respiration/glycolysis, ROS production, oxidative stress, mitochondrial fragmentation and ultrastructure alteration, as observed in FSHD muscle cells. ANT1 overexpression in Xenopus laevis embryos affected skeletal muscle development, impaired skeletal muscle, altered mitochondrial ultrastructure and led to oxidative stress as observed in FSHD muscle biopsies. Moreover, ANT1 overexpression in X. laevis embryos affected heart structure and mitochondrial ultrastructure leading to cardiac arrhythmia, as described in some patients with FSHD. Overall our data suggest that ANT1 could contribute to mitochondria dysfunction and oxidative stress in FSHD muscle cells by modifying their bioenergetic profile associated with ROS production. Such interplay between energy metabolism and ROS production in FSHD will be of significant interest for future prospects. Copyright © 2022 The Authors. Published by Elsevier B.V. All rights reserved. DOI: 10.1016/j.redox.2022.102450 PMCID: PMC9434167 PMID: 36030628 [Indexed for MEDLINE] Conflict of interest statement: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
http://www.ncbi.nlm.nih.gov/pubmed/14659411
1. Neuromuscul Disord. 2004 Jan;14(1):39-45. doi: 10.1016/j.nmd.2003.08.002. Changes in motor cortex excitability in facioscapulohumeral muscular dystrophy. Di Lazzaro V(1), Oliviero A, Tonali PA, Felicetti L, De Marco MB, Saturno E, Pilato F, Pescatori M, Dileone M, Pasqualetti P, Ricci E. Author information: (1)Institute of Neurology, Università Cattolica, Largo A. Gemelli 8, Rome 00168, Italy. vdilazzaro@rm.unicatt.it Previous studies found that some patients with severe, early onset facioscapulohumeral muscular dystrophy (FSHD) present epilepsy and mental retardation. This suggests a functional involvement of central nervous system in severe FSHD. It is unknown whether minor functional changes of central nervous system are also present in less severe forms of FSHD. To investigate this, we examined the excitability of neuronal networks of the motor cortex with a range of transcranial magnetic stimulation paradigms in 20 FSHD patients with heterogeneous clinical severity and compared the data with that from 20 age-matched healthy individuals and from 6 age-matched patients with other muscle diseases. There was significantly less intracortical inhibition in FSHD patients (mean responses +/- SD reduced to 58.1+/-43.5% of the test size) than in controls (mean responses +/- SD reduced to 29.3+/-13.5% of the test size; P=0.025) and in patients with other muscle diseases (mean responses +/-SD, reduced to 30.6+/-11.7% of the test size; P=0.046). No significant difference was found between the control group and patients with other muscle diseases (P=0.970). DOI: 10.1016/j.nmd.2003.08.002 PMID: 14659411 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/23573589
1. Muscle Nerve Suppl. 1995;(2):S67-72. Early onset facioscapulohumeral muscular dystrophy. Brouwer OF(1), Padberg GW, Bakker E, Wijmenga C, Frants RR. Author information: (1)Department of Neurology, Leiden University, Leiden, The Netherlands. We report 10 patients (5 familial, 5 sporadic) with facioscapulohumeral muscular dystrophy (FSHD) with onset of facial and shoulder girdle weakness in early infancy. They showed the same broad range of clinical signs and symptoms as can be seen normally in FSHD. In 7 patients Southern blotting with p13E-11 was performed which showed an abnormal EcoRI fragment (13-22 kb) in 6 of them. We conclude that early onset FSHD does not differ from regular FSHD clinically or genetically. However, the precise mechanisms involved in the extensive clinical variability of the disease are still unknown. PMID: 23573589 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/1908258
1. No To Hattatsu. 1991 Jul;23(4):395-9. [A case of facioscapulohumeral muscular dystrophy with infantile spasms, sensorineural deafness and retinal vessel abnormality]. [Article in Japanese] Akiyama C(1), Suzuki H, Nonaka I. Author information: (1)Department of Pediatrics, Ryokuseikai Seiikuen, Kodaira, Tokyo. We report an 18-year-old female with facioscapulohumeral dystrophy (FSHD), who had sensorineural deafness, retinal vessel abnormality, mental retardation, and epilepsy. She had infantile spasms at 6 months of age. Muscle atrophy and weakness of facial muscles were first noticed at 3 years of age. From 10 years of age, she had rapidly progressive generalized muscle weakness especially of facial, neck and truncal muscles with marked lordosis. Although mental retardation is commonly complicated with FSHD, infantile spasms or epilepsy has never been reported. Not only mental retardation but epilepsy may be one of the central nervous system symptoms in a systemic disorder, FSHD. PMID: 1908258 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35658005
1. N Engl J Med. 2022 Jul 14;387(2):120-131. doi: 10.1056/NEJMoa2204619. Epub 2022 Jun 3. Adagrasib in Non-Small-Cell Lung Cancer Harboring a KRAS(G12C) Mutation. Jänne PA(1), Riely GJ(1), Gadgeel SM(1), Heist RS(1), Ou SI(1), Pacheco JM(1), Johnson ML(1), Sabari JK(1), Leventakos K(1), Yau E(1), Bazhenova L(1), Negrao MV(1), Pennell NA(1), Zhang J(1), Anderes K(1), Der-Torossian H(1), Kheoh T(1), Velastegui K(1), Yan X(1), Christensen JG(1), Chao RC(1), Spira AI(1). Author information: (1)From the Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute (P.A.J.), and Massachusetts General Hospital (R.S.H.) - both in Boston; the Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College (G.J.R.), and Perlmutter Cancer Center, New York University Langone Health (J.K.S.), New York, and the Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo (E.Y.) - all in New York; the Henry Ford Cancer Institute, Detroit (S.M.G.); the University of California Irvine School of Medicine, Chao Family Comprehensive Cancer Center, Orange (S.-H.I.O.),  the University of California San Diego Moores Cancer Center, La Jolla (L.B.), and Mirati Therapeutics, San Diego (K.A., H.D.-T., T.K., K.V., X.Y., J.G.C., R.C.C.) - all in California; the Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora (J.M.P.); Sarah Cannon Research Institute at Tennessee Oncology, Nashville (M.L.J.); the Department of Oncology, Mayo Clinic, Rochester, MN (K.L.); the University of Texas M.D. Anderson Cancer Center, Houston (M.V.N.) and US Oncology Research, The Woodlands (A.I.S.) - both in Texas; Cleveland Clinic Taussig Cancer Institute, Cleveland (N.A.P.); the Division of Medical Oncology, Department of Internal Medicine, and the Department of Cancer Biology, University of Kansas Medical Center, Kansas City (J.Z.); and Virginia Cancer Specialists and NEXT Oncology Virginia - both in Fairfax (A.I.S.). Comment in N Engl J Med. 2022 Jul 14;387(2):180-183. doi: 10.1056/NEJMe2207902. N Engl J Med. 2022 Jul 14;387(2):184-186. doi: 10.1056/NEJMe2206831. Nat Rev Clin Oncol. 2022 Nov;19(11):677-678. doi: 10.1038/s41571-022-00676-4. N Engl J Med. 2022 Sep 29;387(13):1238-1239. doi: 10.1056/NEJMc2210539. BACKGROUND: Adagrasib, a KRASG12C inhibitor, irreversibly and selectively binds KRASG12C, locking it in its inactive state. Adagrasib showed clinical activity and had an acceptable adverse-event profile in the phase 1-1b part of the KRYSTAL-1 phase 1-2 study. METHODS: In a registrational phase 2 cohort, we evaluated adagrasib (600 mg orally twice daily) in patients with KRASG12C -mutated non-small-cell lung cancer (NSCLC) previously treated with platinum-based chemotherapy and anti-programmed death 1 or programmed death ligand 1 therapy. The primary end point was objective response assessed by blinded independent central review. Secondary end points included the duration of response, progression-free survival, overall survival, and safety. RESULTS: As of October 15, 2021, a total of 116 patients with KRASG12C -mutated NSCLC had been treated (median follow-up, 12.9 months); 98.3% had previously received both chemotherapy and immunotherapy. Of 112 patients with measurable disease at baseline, 48 (42.9%) had a confirmed objective response. The median duration of response was 8.5 months (95% confidence interval [CI], 6.2 to 13.8), and the median progression-free survival was 6.5 months (95% CI, 4.7 to 8.4). As of January 15, 2022 (median follow-up, 15.6 months), the median overall survival was 12.6 months (95% CI, 9.2 to 19.2). Among 33 patients with previously treated, stable central nervous system metastases, the intracranial confirmed objective response rate was 33.3% (95% CI, 18.0 to 51.8). Treatment-related adverse events occurred in 97.4% of the patients - grade 1 or 2 in 52.6% and grade 3 or higher in 44.8% (including two grade 5 events) - and resulted in drug discontinuation in 6.9% of patients. CONCLUSIONS: In patients with previously treated KRASG12C -mutated NSCLC, adagrasib showed clinical efficacy without new safety signals. (Funded by Mirati Therapeutics; ClinicalTrials.gov number, NCT03785249.). Copyright © 2022 Massachusetts Medical Society. DOI: 10.1056/NEJMoa2204619 PMID: 35658005 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35666594
1. Cancer Discov. 2022 Aug 5;12(8):OF1. doi: 10.1158/2159-8290.CD-NB2022-0042. Adagrasib Response Remains Strong in NSCLC. [No authors listed] In the phase II KRYSTAL-1 trial, the KRASG12C inhibitor adagrasib yielded an overall response rate of 43% and a disease control rate of 80% in patients with previously treated KRASG12C-mutant non-small cell lung cancer. After a median follow up of 12.9 months, overall survival was 11.7 months. The drug also shrank brain metastases in 33% of patients who had them. ©2022 American Association for Cancer Research. DOI: 10.1158/2159-8290.CD-NB2022-0042 PMID: 35666594 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35608481
1. Clin Cancer Res. 2022 Aug 2;28(15):3179-3181. doi: 10.1158/1078-0432.CCR-22-1137. Trials, Tribunals, and Opportunities for Lung Cancer KRASG12C Brain Metastases. Kommalapati A, Mansfield AS. Comment on Clin Cancer Res. 2022 Aug 2;28(15):3318-3328. doi: 10.1158/1078-0432.CCR-22-0383. The activity of KRAS inhibitors against brain metastases is relatively unexplored. The recent work on preclinical models and preliminary data from the ongoing KRYSTAL-1 phase Ib clinical trial support the potential of adagrasib (MRTX849) to penetrate the central nervous system and provide control of KRASG12C brain metastases. See related article by Sabari et al., p. 3318. ©2022 American Association for Cancer Research. DOI: 10.1158/1078-0432.CCR-22-1137 PMID: 35608481 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35938195
1. J Oncol Pharm Pract. 2023 Mar;29(2):422-430. doi: 10.1177/10781552221118848. Epub 2022 Aug 8. KRAS-targeted therapy in the treatment of non-small cell lung cancer. Yun J(1), Nakagawa R(1), Tham K(1). Author information: (1)Pharmacy, 7284University of Washington/Fred Hutchinson Cancer Center, Seattle, USA. OBJECTIVE: KRAS mutations are one of the most common driver mutations in non-small cell lung cancer. Though previously believed to be an undruggable target, recent advances in therapeutics have seen new targeted agents against KRAS mutations. The objective of this article is to review currently available and upcoming KRAS-targeted treatments. DATA SOURCES: Currently available trials examining KRAS-targeted therapy in non-small cell lung cancer were examined by searching for the keyword "KRAS inhibitors." The pivotal trials for sotorasib and adagrasib were reviewed for this article. DATA SUMMARY: Mutated KRAS can be challenging to target for a variety of reasons. In 2021, the US Food and Drug Administration approved sotorasib for the treatment of adult patients with locally advanced or metastatic non-small cell lung cancer with KRAS G12C mutation as determined by a Food and Drug Administration-approved test, who have received at least one prior systemic therapy. A multicenter, single-group, open-label, phase 2 trial was able to demonstrate that sotorasib was able to demonstrate objective response, progression-free survival, and overall survival in this patient population. A phase 3 trial comparing sotorasib to docetaxel in the subsequent-line treatment of KRAS G12C non-small cell lung cancer is currently ongoing. There are other KRAS-targeted agents currently under study, including adagrasib, with growing interest in targeting KRAS downstream pathways. CONCLUSION: Further trials need to be conducted in order to identify other targeted agents for KRAS and the appropriate place in therapy among currently approved treatments for non-small cell lung cancer. DOI: 10.1177/10781552221118848 PMID: 35938195 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/15189359
1. J Neurochem. 2004 Jun;89(6):1555-7. doi: 10.1111/j.1471-4159.2004.02544.x. Endocannabinoids accumulate in spinal cord of SOD1 G93A transgenic mice. Witting A(1), Weydt P, Hong S, Kliot M, Moller T, Stella N. Author information: (1)Department of Pharmacology, University of Washington, Seattle, WA, USA. Approximately 2% of amyotrophic lateral sclerosis (ALS) cases are caused by mutations in the super oxide dismutase 1 (SOD1) gene and transgenic mice for these mutations recapitulate many features of this devastating neurodegenerative disease. Here we show that the amount of anandamide (AEA) and 2-arachidonoylglycerol (2-AG), two endocannabinoids that have neuroprotective properties, increase in spinal cord of SOD1(G93A) transgenic mice. This increase occurs in the lumbar section of spinal cords, the first section to undergo neurodegeneration, and is significant before overt motor impairment. Our results show that chronic neurodegeneration induced by a genetic mutation increases endocannabinoid production possibly as part of an endogenous defense mechanism. DOI: 10.1111/j.1471-4159.2004.02544.x PMID: 15189359 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/30509290
1. Mol Neurodegener. 2018 Dec 4;13(1):63. doi: 10.1186/s13024-018-0294-0. Disease-modifying effects of metabolic perturbations in ALS/FTLD. Jawaid A(1)(2), Khan R(3), Polymenidou M(4), Schulz PE(5). Author information: (1)Laboratory of Neuroepigenetics, Brain Research Institute, University of Zurich (UZH)/ Swiss Federal Institute of Technology (ETH), Winterthurerstr. 190, 8057, Zurich, Switzerland. jawaid@hifo.uzh.ch. (2)Syed Babar Ali School of Science and Engineering (SBASSE), Lahore University of Management Sciences (LUMS), Lahore, Pakistan. jawaid@hifo.uzh.ch. (3)Syed Babar Ali School of Science and Engineering (SBASSE), Lahore University of Management Sciences (LUMS), Lahore, Pakistan. (4)Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland. (5)Department of Neurology, The McGovern Medical School of UT Health, Houston, TX, USA. Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) are two fatal neurodegenerative disorders with considerable clinical, pathological and genetic overlap. Both disorders are characterized by the accumulation of pathological protein aggregates that contain a number of proteins, most notably TAR DNA binding protein 43 kDa (TDP-43). Surprisingly, recent clinical studies suggest that dyslipidemia, high body mass index, and type 2 diabetes mellitus are associated with better clinical outcomes in ALS. Moreover, ALS and FTLD patients have a significantly lower incidence of cardiovascular disease, supporting the idea that an unfavorable metabolic profile may be beneficial in ALS and FTLD. The two most widely studied ALS/FTLD models, super-oxide dismutase 1 (SOD1) and TAR DNA binding protein of 43 kDA (TDP-43), reveal metabolic dysfunction and a positive effect of metabolic strategies on disease onset and/or progression. In addition, molecular studies reveal a role for ALS/FTLD-associated proteins in the regulation of cellular and whole-body metabolism. Here, we systematically evaluate these observations and discuss how changes in cellular glucose/lipid metabolism may result in abnormal protein aggregations in ALS and FTLD, which may have important implications for new treatment strategies for ALS/FTLD and possibly other neurodegenerative conditions. DOI: 10.1186/s13024-018-0294-0 PMCID: PMC6278047 PMID: 30509290 [Indexed for MEDLINE] Conflict of interest statement: ETHICS APPROVAL AND CONSENT TO PARTICIPATE: N/A. CONSENT FOR PUBLICATION: The authors willfully consent for publication of this article. COMPETING INTERESTS: The authors declare that they have no competing interests. PUBLISHER’S NOTE: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
http://www.ncbi.nlm.nih.gov/pubmed/12402272
1. Ann Neurol. 2002 Nov;52(5):680-3. doi: 10.1002/ana.10369. "True" sporadic ALS associated with a novel SOD-1 mutation. Alexander MD(1), Traynor BJ, Miller N, Corr B, Frost E, McQuaid S, Brett FM, Green A, Hardiman O. Author information: (1)Department of Neurology, Beaumont Hospital, Beaumont Road, Dublin 9, Ireland. mikealexan@hotmail.com Mutations in the Cu/Zn superoxide dismutase gene (SOD-1) are reported in 20% of familial amyotrophic lateral sclerosis (ALS) cases, but no definite report of a mutation in a "truly" sporadic case of ALS has been proved. We present the first case of a novel SOD-1 mutation in a patient with genetically proven sporadic ALS. This mutation (H80A) is believed to alter zinc ligand binding, and its functional significance correlates well with the aggressive clinical course and postmortem findings observed in this patient. DOI: 10.1002/ana.10369 PMID: 12402272 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/10624810
1. Neurosci Lett. 1999 Dec 3;276(2):135-7. doi: 10.1016/s0304-3940(99)00803-4. A novel mutation (Cys6Gly) in the Cu/Zn superoxide dismutase gene associated with rapidly progressive familial amyotrophic lateral sclerosis. Kohno S(1), Takahashi Y, Miyajima H, Serizawa M, Mizoguchi K. Author information: (1)The First Department of Medicine, Hamamatsu University School of Medicine, Japan. sato-kohno@mail.wbs.ne.jp Autosomal-dominant familial amyotrophic lateral sclerosis (FALS) is associated with mutation in the gene that encodes Cu/Zn superoxide dismutase (SOD1). We identified a novel missense mutation of SOD-1 (Cys6Gly) in exon 1 in a Japanese woman and her family. The illness showed rapid progression similarly to the FALS with a mutation of Cys6Phe that was reported by Morita et al. (1996) (Morita, M., Aoki, M., Abe, K., Hasegawa, T., Sakuma, R., Onodera, Y., Ichikawa, N., Nishizawa, M. and Itoyama, Y., A novel two-base mutation in the Cu/Zn superoxide dismutase gene associated with familial amyotrophic lateral sclerosis in Japan. Neurosci. Lett., 205 (1996) 79-82). Mutation of the cystein at amino acid 6 might be associated with the rapid progression of ALS. DOI: 10.1016/s0304-3940(99)00803-4 PMID: 10624810 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/24283821
1. Eur J Neurol. 1997 Jan;4(1):48-51. doi: 10.1111/j.1468-1331.1997.tb00298.x. A novel mutation of SOD-1 (Gly 108 Val) in familial amyotrophic lateral sclerosis. Orrell RW(1), Habgood JJ, Shepherd DI, Donnai D, de Belleroche J. Author information: (1)Department of Biochemistry, Charing Cross and Westminster Medical School, LondonNeuromuscular Unit, Charing Cross Hospital, LondonDepartment of Neurology, North Manchester General Hospital, ManchesterRegional Genetic Service, St Mary's Hospital, Manchester, UK. A novel mutation of the SOD-1 gene which encodes the enzyme copper-zinc superoxide dismutase was identified in a family manifesting amyotrophic lateral sclerosis (ALS) in three generations. The mutation is a heterozygote point mutation in exon 4, codon 108 (GGA to GTA), predicting the substitution of valine for glycine. The mutation creates a new restriction site for the endonuclease AccI. The mutation was demonstrated in two affected members of the family, who show features of autosomal dominant inheritance of ALS, but variable age at onset ranging from 48 to 72 years. Over 30 different mutations of SOD-1 have now been identified in families with ALS. The definition of the different mutations causing human disease may allow further investigation of their pathogenicity in transgenic animal models, and also offers insight into the variable phenotypic disease expression both within and between genotypes. 1997 Lippincott Williams & Wilkins. DOI: 10.1111/j.1468-1331.1997.tb00298.x PMID: 24283821
http://www.ncbi.nlm.nih.gov/pubmed/11284995
1. Eur J Neurol. 2001 Mar;8(2):167-72. doi: 10.1046/j.1468-1331.2001.00186.x. Sporadic ALS associated with the D90A Cu,Zn superoxide dismutase mutation in Russia. Skvortsova VI(1), Limborska SA, Slominsky PA, Levitskaya NI, Levitsky GN, Shadrina MI, Kondratyeva EA. Author information: (1)Department of Neurology with Laboratory and Functional Diagnostics of Russian State Medical University, Moscow. vskvor@sl.ru Twenty blood samples from Russian patients (Moscow) with idiopathic motor neurone disease were analysed for mutations in the Cu,Zn superoxide dismutase (Cu,Zn SOD) gene. Two patients (10%) with the amyotrophic lateral sclerosis (ALS) form of the disease were found to have a disease-related mutation. One patient appears to have autosomal recessive adult-onset ALS associated with homozygosity for D90A and presents the characteristic phenotype of very slowly ascending paresis with both lower and upper motor neurone signs. Another patient, heterozygous for D90A, presents ALS with lumbar onset and rapid progression. This is the first report of a Cu,Zn SOD mutation in ALS in Russia. DOI: 10.1046/j.1468-1331.2001.00186.x PMID: 11284995 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/10202988
1. J Neurol Sci. 1999 Jan 15;162(2):201-4. doi: 10.1016/s0022-510x(98)00237-8. Heterogeneous distribution of amyotrophic lateral sclerosis patients with SOD-1 gene mutations: preliminary data on an Italian survey. Malaspina A(1), Zaman R, Mazzini L, Camana C, Poloni E, Curti D, Ceroni M. Author information: (1)Institute of Neurology, Foundation Casimiro Mondino, Laboratory of Neurogenetics, University of Pavia, Italy. rabc035@s1.cxwms.ac.uk We report the absence of superoxide dismutase (SOD-1) gene mutations in 30 patients with amyotrophic lateral sclerosis (ALS) including individuals with a confirmed family history of ALS (familial ALS/FALS), ALS with an unclear family history (UFALS) and sporadic ALS (SALS). Single strand conformation polymorphism (SSCP) and sequence analysis of the 5 SOD-1 gene exons were undertaken to improve the accuracy of the mutation detection. Our preliminary data appear to diverge from the results of studies by other groups using different populations. We discuss the possible reasons for this disparity and the apparent heterogeneous distribution of ALS with SOD-1 gene mutations among different ethnic groups. DOI: 10.1016/s0022-510x(98)00237-8 PMID: 10202988 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/20385392
1. J Neurol Sci. 2010 Jun 15;293(1-2):112-5. doi: 10.1016/j.jns.2010.03.009. Epub 2010 Apr 10. Severe familial ALS with a novel exon 4 mutation (L106F) in the SOD1 gene. Battistini S(1), Ricci C, Lotti EM, Benigni M, Gagliardi S, Zucco R, Bondavalli M, Marcello N, Ceroni M, Cereda C. Author information: (1)Department of Neuroscience, University of Siena, Siena, Italy. battistinis@unisi.it Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disease associated with a positive familial history in 5-10% of ALS cases. Mutations in the superoxide dismutase-1 (SOD1) gene have been found in 12%-23% of patients diagnosed with familial ALS. Here we report a novel mutation in exon 4 of SOD1 gene in a 55-year-old ALS patient belonging to a large Italian family with ALS first clinically described in 1968. In the family the clinical presentation was characterized by relatively early age of onset, spinal onset with proximal distribution weakness, bulbar involvement and a rapid disease course. Molecular analysis showed a heterozygous mutation at codon 106 resulting in a substitution of phenylalanine for leucine in the SOD1 protein (L106F). In analogy with the previously reported L106V mutation, we propose that the L106F causes a relevant destabilization of the protein chain around the mutation site, able to affect the SOD1 monomer and dimer structures suggesting a pathogenic role for this novel mutation. Copyright 2010 Elsevier B.V. All rights reserved. DOI: 10.1016/j.jns.2010.03.009 PMID: 20385392 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/9455977
1. J Neurol Sci. 1997 Dec 9;153(1):46-9. doi: 10.1016/s0022-510x(97)00181-0. Familial ALS is associated with mutations in all exons of SOD1: a novel mutation in exon 3 (Gly72Ser). Orrell RW(1), Marklund SL, deBelleroche JS. Author information: (1)Department of Biochemistry, Charing Cross and Westminster Medical School, Charing Cross Hospital, London, UK. rorrell@mail.neurology.rochester.edu Mutations of the SOD1 gene, which encodes the enzyme copper/zinc superoxide dismutase, are associated with familial amyotrophic lateral sclerosis (ALS). SOD1 consists of five exons, and over 50 different mutations have been described involving exons 1,2,4 and 5. The absence of mutations in exon 3 has been attributed to a critical function of this exon, its integrity being necessary for the toxic effect of mutant SOD1, and it has been suggested that such mutations may be lethal rather than leading to adult onset disease. We identified the heterozygote mutation Gly72Ser (exon 3) in a family with two individuals affected by ALS. SOD enzyme activity was reduced by 45% when measured in erythrocytes indicating reduced enzyme activity, or reduced stability of the mutant protein. These findings indicate that exon 3 is not a privileged region from mutation; that all five exons should be investigated when seeking SOD1 mutations in human disease; and may help in a better understanding of the pathogenicity of these mutations in ALS. DOI: 10.1016/s0022-510x(97)00181-0 PMID: 9455977 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/27297615
1. Neurobiol Aging. 2016 Sep;45:212.e1-212.e4. doi: 10.1016/j.neurobiolaging.2016.04.021. Epub 2016 May 3. A novel 10-base pair insertion mutation in exon 5 of the SOD1 gene in a Chinese family with amyotrophic lateral sclerosis. Chen S(1), Li M(1), Zhu W(1), Mao F(2), Wang J(2), Sun Z(3), Huang X(4). Author information: (1)Department of Neurology, Chinese PLA General Hospital, Beijing, China. (2)Beijing Institutes of Life Science Chinese Academy of Science, Beijing, China; University of Chinese Academy of Science, Beijing, China. (3)Beijing Institutes of Life Science Chinese Academy of Science, Beijing, China; University of Chinese Academy of Science, Beijing, China. Electronic address: zsunusa@yahoo.com. (4)Department of Neurology, Chinese PLA General Hospital, Beijing, China. Electronic address: lewish301@sina.com. Amyotrophic lateral sclerosis (ALS) is an adult-onset, progressive, fatal neurodegenerative disease. Several genes are associated with ALS. Copper-zinc superoxide dismutase 1 (SOD1) is one of the most commonly mutated genes in ALS, and more than 160 mutations in SOD1 have been reported. We reported a novel heterozygous insertion mutation that led to a frameshift and a premature termination at position 136 in exon 5 of the SOD1 gene (c.392_393insGCAAAGGTGG; p.N132Qfs*5) in a Chinese familial ALS pedigree. This mutation in the pedigree demonstrated an autosomal dominant pattern of inheritance and a phenotype characterized by an early onset (approximately 34 years old) with a relatively rapid course (approximately 2 years) and limb onset with respiratory involvement. The clinical feature of the p.N132Qfs*5 mutation was nearly identical to a previously reported mutation (Gly127insTGGG). Experiments in G127X mice demonstrated that the G127X mutation was pathogenic. SOD1 activity in the p.N132Qfs*5 mutation carriers in the family decreased significantly compared with normal family members. In conclusion, we identified a novel SOD1 mutation in an ALS family, which is an important addition to the catalog of SOD1 mutations in ALS. Copyright © 2016 Elsevier Inc. All rights reserved. DOI: 10.1016/j.neurobiolaging.2016.04.021 PMID: 27297615 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/7951249
1. Hum Mol Genet. 1994 Jun;3(6):981-7. doi: 10.1093/hmg/3.6.981. A frequent ala 4 to val superoxide dismutase-1 mutation is associated with a rapidly progressive familial amyotrophic lateral sclerosis. Rosen DR(1), Bowling AC, Patterson D, Usdin TB, Sapp P, Mezey E, McKenna-Yasek D, O'Regan J, Rahmani Z, Ferrante RJ, et al. Author information: (1)Day Neuromuscular Research Laboratory, Massachusetts General Hospital, Charlestown 02129-2060. Familial amyotrophic lateral sclerosis (FALS), a degenerative disorder of motor neurons, is associated with mutations in the Cu/Zn superoxide dismutase gene SOD1 in some affected families. We confirm a recently reported ala4-->val mutation in exon 1 of the SOD1 gene and report that this mutation is both the most commonly detected of all SOD1 mutations and among the most clinically severe. By comparison with our other FALS families, the exon 1 mutation is associated with reduced survival time after onset: 1.2 years, as compared to 2.5 years for all other FALS patients. We also demonstrate that SOD1 is prominently expressed in normal motor neurons and that neural expression of SOD1 is not prevented by this exon 1 mutation. Assays of SOD1 enzymatic activity in extracts from red blood cells, lymphoblastoid cells, and brain tissues revealed an approximately 50% reduction in activity of cytosolic SOD1 in patients with this mutation compared to normal individuals. By contrast, patients with sporadic ALS had normal levels of SOD1 enzymatic activity. Why this SOD1 mutation causes motor neuron death in FALS remains to be established. While it may be that FALS is a consequence of loss of SOD1 function, it is also possible that motor neuron death in this dominantly inherited disease occurs because the mutations confer an additional, cytotoxic function on the SOD1 protein. DOI: 10.1093/hmg/3.6.981 PMID: 7951249 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/7820674
1. Brain Res. 1994 Oct 3;659(1-2):272-6. doi: 10.1016/0006-8993(94)90892-3. Copper/zinc superoxide dismutase mRNA levels are increased in sporadic amyotrophic lateral sclerosis motorneurons. Bergeron C(1), Muntasser S, Somerville MJ, Weyer L, Percy ME. Author information: (1)Centre for Research in Neurodegenerative Diseases, University of Toronto, Ont., Canada. Mutations of the Cu/Zn superoxide dismutase (SOD-1) gene were recently implicated in the pathogenesis of familial amyotrophic lateral sclerosis (ALS). We measured SOD-1 mRNA levels in motorneurons of the more common sporadic form of the disease and found a 42% increase in ALS motorneurons (P = 0.058) as compared with controls. These results suggest that oxidative stress may also play a role in the pathogenesis of sporadic ALS. DOI: 10.1016/0006-8993(94)90892-3 PMID: 7820674 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/9065559
1. Neurology. 1997 Mar;48(3):746-51. doi: 10.1212/wnl.48.3.746. Clinical and functional investigation of 10 missense mutations and a novel frameshift insertion mutation of the gene for copper-zinc superoxide dismutase in UK families with amyotrophic lateral sclerosis. Orrell RW(1), Habgood JJ, Gardiner I, King AW, Bowe FA, Hallewell RA, Marklund SL, Greenwood J, Lane RJ, deBelleroche J. Author information: (1)Department of Biochemistry, Charing Cross and Westminster Medical School, London, England. Mutations of the gene SOD-1, which encodes the enzyme copper-zinc superoxide dismutase, occur in patients with a familial form of amyotrophic lateral sclerosis (ALS). We investigated 71 families with more than one individual affected by ALS for clinical features and SOD-1 mutations. Mutations were identified in 14 families, indicating the presence of SOD-1 mutations in around 20% of this population. There were 10 different heterozygote missense point mutations in eight different codons, and a novel two-base frameshift insertion (132insTT), which leads to substitution of aspartic acid for glutamic acid at codon 132, and a premature stop codon at 133, with predicted truncation of the protein. SOD enzyme activity was reduced to around 50% of normal in individuals with SOD-1 mutations, and may be a useful predictor for the presence of these mutations. A predilection for disease onset in the lower limbs appears to be a distinguishing feature of familial ALS with SOD-1 mutations, and accords with findings in transgenic mouse models. In general, the finding of an SOD-1 mutation does not accurately predict a prognosis or disease severity. DOI: 10.1212/wnl.48.3.746 PMID: 9065559 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/9506558
1. Ann Neurol. 1998 Mar;43(3):390-4. doi: 10.1002/ana.410430319. Mutations in all five exons of SOD-1 may cause ALS. Shaw CE(1), Enayat ZE, Chioza BA, Al-Chalabi A, Radunovic A, Powell JF, Leigh PN. Author information: (1)Department of Clinical Neurosciences, Institute of Psychiatry and King's College School of Medicine and Dentistry, London, United Kingdom. Eight of 38 patients (21%) with familial and 5 of 175 patients (3%) with sporadic amyotrophic lateral sclerosis (ALS) had missense mutations in the SOD-1 gene. Two novel mutations were identified. One in exon 4 substituting leucine with phenylalanine (L84F) in a familial patient and the second in exon 3 at substituting glycine with serine (G72S) in an "apparently" sporadic patient. Over 60 point mutations have now been described in all five exons of SOD-1, involving 43 of the 153 residues. Hypotheses about the toxic role of mutant SOD-1 in the pathogenesis of ALS must account for this molecular diversity. DOI: 10.1002/ana.410430319 PMID: 9506558 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/7917302
1. Neuron. 1994 Sep;13(3):727-36. doi: 10.1016/0896-6273(94)90039-6. Analysis of the functional effects of a mutation in SOD1 associated with familial amyotrophic lateral sclerosis. Tsuda T(1), Munthasser S, Fraser PE, Percy ME, Rainero I, Vaula G, Pinessi L, Bergamini L, Vignocchi G, McLachlan DR, et al. Author information: (1)Department of Medicine, University of Toronto, Toronto, Ontario, Canada. Mutations in the Cu, Zn superoxide dismutase (SOD1) gene have been reported in some pedigrees with Familial Amyotrophic Lateral Sclerosis (FALS). We have investigated the functional and structural effects of a Gly-->Ser mutation at codon 41 of SOD1 in a pedigree with FALS and the topography of SOD1 expression in the mammalian CNS. These analyses show that the 41Gly-->Ser mutation causes a 27% reduction in Cu, Zn SOD activity. SOD1 is transcribed at high levels in rat motoneurons and four other types of neurons homologous to upper motoneurons that degenerate in human ALS. However, SOD1 is transcribed at lower levels in other types of neurons, such as cerebellar Purkinje cells, which are not usually involved significantly in human ALS. On the other hand, immunocytochemical studies indicate that most types of rat neurons contain similar levels of Cu, Zn SOD immunoreactive protein. Nevertheless, these results suggest that the essential feature causing this subtype of ALS is either a reduction in Cu, Zn SOD activity in cell types that presumably critically require Cu, Zn SOD for protection against oxidative damage or the fact that the mutation in SOD1 associated with FALS results in a novel gain of function that is particularly deleterious to those cell types expressing SOD1 at high levels. DOI: 10.1016/0896-6273(94)90039-6 PMID: 7917302 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/11464949
1. Amyotroph Lateral Scler Other Motor Neuron Disord. 2000 Jun;1(3):143-61. doi: 10.1080/14660820050515151. Superoxide dismutase-1 mutation-related neurotoxicity in familial amyotrophic lateral sclerosis. Shibata N(1), Hirano A, Yamamoto T, Kato Y, Kobayashi M. Author information: (1)Department of Pathology, Tokyo Women's Medical University, Japan. shibatan@research.twmu.ac.jp Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder characterized by motor neuron system involvement, and is epidemiologically subclassified into sporadic, familial and endemic forms. About 20% of ALS families are associated with mutations in the gene for superoxide dismutase-1 (SOD1) encoded on chromosome 21q22.1. Several studies have pointed to a variety of functions of mutant SOD1, which has enhanced catalytic activity of the peroxynitrite-mediated tyrosine nitration, readily releases the reactive Cu ions, induces apoptotic cell death, has enhanced peroxidase activity, damages the mitochondria to release Ca2+, and forms SOD1-containing aggregates in the cytoplasm. Many of these studies have obtained evidence for increased oxidative damage in ALS. On the other hand, some reports disagree with oxidative damage involvement in SOD1 mutant ALS. In considering the findings of increased oxidative damage in mutant SOD1-expressing transgenic mice, it should be remembered that overexpression of mutant SOD1 may enhance oxidative stress generation from this enzyme. In this review, we present the clinicopathological features of SOD1 mutant familial ALS and its transgenic mouse model, and also discuss SOD1 mutation-related neurotoxicity, including SOD1 protein aggregation and post-translational protein modification. DOI: 10.1080/14660820050515151 PMID: 11464949 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/8446170
1. Nature. 1993 Mar 4;362(6415):59-62. doi: 10.1038/362059a0. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Rosen DR(1), Siddique T, Patterson D, Figlewicz DA, Sapp P, Hentati A, Donaldson D, Goto J, O'Regan JP, Deng HX, et al. Author information: (1)Day Neuromuscular Research Laboratory, Massachusetts General Hospital, Charlestown 02129. Erratum in Nature. 1993 Jul 22;364(6435):362. doi: 10.1038/364362c0. Comment in Nature. 1993 Mar 4;362(6415):20-1. doi: 10.1038/362020a0. Amyotrophic lateral sclerosis (ALS) is a degenerative disorder of motor neurons in the cortex, brainstem and spinal cord. Its cause is unknown and it is uniformly fatal, typically within five years. About 10% of cases are inherited as an autosomal dominant trait, with high penetrance after the sixth decade. In most instances, sporadic and autosomal dominant familial ALS (FALS) are clinically similar. We have previously shown that in some but not all FALS pedigrees the disease is linked to a genetic defect on chromosome 21q (refs 8, 9). Here we report tight genetic linkage between FALS and a gene that encodes a cytosolic, Cu/Zn-binding superoxide dismutase (SOD1), a homodimeric metalloenzyme that catalyzes the dismutation of the toxic superoxide anion O2.- to O2 and H2O2 (ref. 10). Given this linkage and the potential role of free radical toxicity in other neurodenegerative disorders, we investigated SOD1 as a candidate gene in FALS. We identified 11 different SOD1 missense mutations in 13 different FALS families. DOI: 10.1038/362059a0 PMID: 8446170 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/23954173
1. Neurobiol Aging. 2014 Jan;35(1):266.e1-4. doi: 10.1016/j.neurobiolaging.2013.07.012. Epub 2013 Aug 15. Novel SOD1 mutation p.V31A identified with a slowly progressive form of amyotrophic lateral sclerosis. Dangoumau A(1), Verschueren A, Hammouche E, Papon MA, Blasco H, Cherpi-Antar C, Pouget J, Corcia P, Andres CR, Vourc'h P. Author information: (1)UMR INSERM U930, Université François-Rabelais, Tours, PRES Centre - Val de Loire Université, France. The SOD1 gene encoding the superoxide dismutase 1 (SOD1) protein is mutated in approximately 15% of familial amyotrophic lateral sclerosis (ALS) and 3% of sporadic ALS. We identified a novel mutation in SOD1 in a man who presented at age 49 with lower limb stiffness, and at age 53, a spastic paraparesia with distal muscular atrophy in the lower limbs and fasciculations in the quadriceps. A diagnosis of ALS was established. Eleven years after disease onset his condition continues gradually and slowly to deteriorate. The heterozygous mutation observed in exon 2 resulted in a valine to alanine substitution at position 31 in the β-barrel domain of the SOD1 protein. Functional analysis in NSC34 cells showed that the overexpression of the mutant form of SOD1(V31A) induced aggregates and decreased cell viability. This mutation is located outside of the regions carrying most of the ALS-related mutations (i.e., the catalytic center, the region of dimerization, and the loops between the β-strands of the β-barrel). In conclusion, we identified a novel SOD1 mutation in a patient with slow disease progression and supported the idea that different SOD1 mutations can lead to distinct ALS phenotypes. Copyright © 2013 Elsevier Inc. All rights reserved. DOI: 10.1016/j.neurobiolaging.2013.07.012 PMID: 23954173 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/27556028
1. Front Mol Biosci. 2016 Aug 9;3:40. doi: 10.3389/fmolb.2016.00040. eCollection 2016. Screening of Drugs Inhibiting In vitro Oligomerization of Cu/Zn-Superoxide Dismutase with a Mutation Causing Amyotrophic Lateral Sclerosis. Anzai I(1), Toichi K(1), Tokuda E(1), Mukaiyama A(2), Akiyama S(2), Furukawa Y(1). Author information: (1)Laboratory for Mechanistic Chemistry of Biomolecules, Department of Chemistry, Keio University Yokohama, Japan. (2)Research Center of Integrative Molecular Systems, Institute for Molecular ScienceOkazaki, Japan; Department of Functional Molecular Science, SOKENDAI (The Graduate University for Advanced Studies)Okazaki, Japan. Dominant mutations in Cu/Zn-superoxide dismutase (SOD1) gene have been shown to cause a familial form of amyotrophic lateral sclerosis (SOD1-ALS). A major pathological hallmark of this disease is abnormal accumulation of mutant SOD1 oligomers in the affected spinal motor neurons. While no effective therapeutics for SOD1-ALS is currently available, SOD1 oligomerization will be a good target for developing cures of this disease. Recently, we have reproduced the formation of SOD1 oligomers abnormally cross-linked via disulfide bonds in a test tube. Using our in vitro model of SOD1 oligomerization, therefore, we screened 640 FDA-approved drugs for inhibiting the oligomerization of SOD1 proteins, and three effective classes of chemical compounds were identified. Those hit compounds will provide valuable information on the chemical structures for developing a novel drug candidate suppressing the abnormal oligomerization of mutant SOD1 and possibly curing the disease. DOI: 10.3389/fmolb.2016.00040 PMCID: PMC4977284 PMID: 27556028
http://www.ncbi.nlm.nih.gov/pubmed/15679043
1. Folia Neuropathol. 2004;42(4):239-48. Transgenic models of amyotrophic lateral sclerosis. Grieb P(1). Author information: (1)Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warszawa, Poland. pgrieb@cmdik.pan.pl Amyotrophic lateral sclerosis (ALS), the most frequent paralytic disease of adults, is untreatable and invariably fatal. Up to 20% of ALS cases are inherited (familial, fALS) and associated with mutations, usually of the superoxide dismutase type 1 (SOD-1) gene. This paper shortly reviews the background for and the use of rodent transgenic models of ALS. Silencing the SOD-1 gene does not produce paralytic phenotype, but transgenic rodents expressing human mutated (hm) SOD-1 atop their own enzyme develop relatively selective and fatal degeneration of motoneurons. Many essential neuropathological and biochemical features of the paralytic disease in hmSOD-1 transgenic mice and rats are similar to human fALS, and these animals are currently considered a model of the human disease. Two types of hypotheses put forward to explain pathomechanism of motoneuron degeneration in hmSOD-1 transgenics (hence also in human ALS) are the "gain of function" hypotheses which assume that the mutated enzyme displays new toxic catalytic properties, and the "gain of interaction" hypotheses which assume that the mutated protein molecules are toxic because they became misfolded and undergo oligomerization. Transgenic animal models of ALS are used for preclinical assessments of new therapeutic approaches ranging from mono- and polipharmacotherapy to gene therapy and stem cell therapy. PMID: 15679043 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/15079798
1. Zhonghua Yi Xue Yi Chuan Xue Za Zhi. 2004 Apr;21(2):149-52. [Identification of the mutation of SOD1 gene in a familial amyotrophic lateral sclerosis]. [Article in Chinese] Shi SG(1), Li LS, Chen KN, Liu X. Author information: (1)Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing, 400038 PR China. shishg@mail.tmmu.com.cn OBJECTIVE: To identify the mutation of Cu/Zn superoxide dismutase(SOD1) gene in an amyotrophic lateral sclerosis (ALS) family with unique phenotype. METHODS: Five exons of SOD1 gene were amplified by PCR. The differences of these products were analyzed by PCR-single strand conformation polymorphism and visualized by silver staining. RESULTS: Abnormal bands were found in exons 2 and 5 of SOD1 gene in several familial members. DNA sequence analysis verified that a base pair insertion occurred in the codon area of exon 2 and in the intron area of exon 5. And the insertion mutation of exon 2 led to a frameshift mutation and premature stop. It is a new type of SOD1 mutation which may be associated with familial amyotrophic lateral sclerosis. CONCLUSION: Insertion mutation of exon 2 may be responsible for the disease of an ALS family in Chongqing. PMID: 15079798 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/31928838
1. Bioorg Med Chem Lett. 2020 Feb 15;30(4):126950. doi: 10.1016/j.bmcl.2020.126950. Epub 2020 Jan 7. Development of novel small molecules for the treatment of ALS. Mathew B(1), Ruiz P(1), Pathak V(1), Suto MJ(2). Author information: (1)Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL 35205, USA. (2)Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL 35205, USA. Electronic address: msuto@southernresearch.org. Amyotrophic lateral sclerosis (ALS) is a rare and progressive neurodegenerative disease with unknown etiology. It is caused by the degeneration of motor neurons responsible for controlling voluntary muscles. It has been reported that mutations in the superoxide dismutase (SOD) 1 gene can lead to ALS. SOD1 abnormalities have been identified in both familial, as well as sporadic ALS cases. SOD2 is a highly inducible SOD that works in conjunction with SOD1. SOD2 can be induced through activation of NF-κBs. We previously reported that the novel small molecule, SRI-22818, increases NF-κB expression and activation and SOD2 levels in vitro and has activity in vivo in the SOD1-G93A reference model of ALS. We report herein the synthesis and biological evaluation of SRI-22818 analogs. Copyright © 2020 Elsevier Ltd. All rights reserved. DOI: 10.1016/j.bmcl.2020.126950 PMID: 31928838 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/10869061
1. Brain. 2000 Jul;123 ( Pt 7):1505-15. doi: 10.1093/brain/123.7.1505. Preserved slow conducting corticomotoneuronal projections in amyotrophic lateral sclerosis with autosomal recessive D90A CuZn-superoxide dismutase mutation. Weber M(1), Eisen A, Stewart HG, Andersen PM. Author information: (1)The Neuromuscular Diseases Unit, Vancouver Hospital and the University of British Columbia, Vancouver, Canada. Recently, a subgroup of the amyotrophic lateral sclerosis (ALS) syndrome associated with mutations in the gene encoding the free radical scavenging enzyme CuZn-superoxide dismutase (CuZn-SOD, SOD1) has been identified. Some 67 different mutations have been reported worldwide to date, comprising about one-fifth of familial ALS cases in the populations studied. The autosomal recessively inherited D90A CuZn-SOD mutation has been associated with a very slowly progressive, clinically distinct phenotype, and is neurophysiologically characterized by very slow central motor conduction. It is not known which physiological and/or biochemical mechanisms are responsible for the different clinical course. To delineate ALS associated with this particular CuZn-SOD mutation from ALS without mutations, we performed a detailed neurophysiological study of the corticomotoneuronal function using peristimulus time histograms (PSTHs) in eight ALS patients homozygous for the D90A CuZn-SOD mutation. The results were compared with those obtained in 12 non-hereditary ALS patients and 11 healthy subjects. PSTHs were constructed from three to seven different, voluntarily recruited motor units of the extensor digitorum communis muscle (EDC) in each patient. The onset latency, number of excess bins, duration and synchrony of the primary peak were analysed. All measurements differed significantly between healthy controls and the D90A patients (P < 0.0007). The mean onset latency of the primary peak in D90A patients was 35.3 ms, compared with 24.2 ms for non-hereditary ALS patients and 19.3 ms for normal subjects (P < 0.0000). Delayed primary peaks in the D90A patients were desynchronized and characteristically preceded by a marked suppression phase. This suppression phase was not seen in non-hereditary ALS patients. We conclude that the mainly slow conducting and/or polysynaptic corticomotoneuronal connections are preserved in the D90A homozygous cases, and that the cortical and possibly spinal inhibitory circuitry is preserved. These events may partially protect the motor neurons, slowing down the degenerative process. DOI: 10.1093/brain/123.7.1505 PMID: 10869061 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/8981308
1. J Neurol Sci. 1996 Nov;143(1-2):118-20. doi: 10.1016/s0022-510x(96)00190-6. Serum Cu/Zn superoxide dismutase activity is reduced in sporadic amyotrophic lateral sclerosis patients. Cohen O(1), Kohen R, Lavon E, Abramsky O, Steiner I. Author information: (1)Department of Neurology, Hadassah University Hospital, Hebrew University Hadassah Medical School, Jerusalem, Israel. Comment in J Neurol Sci. 1997 Nov 25;152(2):226, 229. Accumulated evidence implies that mutations in the gene coding for Cu/Zn superoxide dismutase (SOD) are associated with the pathogenesis of the familial form of amyotrophic lateral sclerosis (ALS). The clinical and pathological similarities of the familial and the sporadic forms of the disease may suggest that abnormal SOD activity takes also part in the pathogenesis of sporadic ALS. We therefore measured serum SOD activity in fifteen sporadic ALS patients. Mean serum SOD activity was 1.15 +/- 0.40 u/ml in ALS patients, 1.50 +/- 0.45 u/ml. in patients with other neurological disorders and 1.45 +/- 0.45 u/ml in.healthy controls (p < 0.021 and p < 0.031 respectively). If this sporadic ALS-related reduction in serum SOD activity will be confirmed in the diseased nervous system tissue, it may suggest that abnormal SOD activity is also associated with the motor neuron damage in the sporadic form of ALS. DOI: 10.1016/s0022-510x(96)00190-6 PMID: 8981308 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/24611504
1. Amyotroph Lateral Scler Frontotemporal Degener. 2014 Jun;15(3-4):312-4. doi: 10.3109/21678421.2013.873051. Epub 2014 Mar 10. Multiple system involvement in a Japanese patient with a V31A mutation in the SOD1 gene. Sakamoto H(1), Akamatsu M, Hirano M, Saigoh K, Ueno S, Isono C, Kusunoki S, Nakamura Y. Author information: (1)Department of Neurology, Sakai Hospital Kinki University Faculty of Medicine , Osaka , Japan. The superoxide dismutase-1 (SOD1) gene is the first gene for familial amyotrophic lateral sclerosis (ALS) with autosomal dominant inheritance. We describe a Japanese patient who had slowly progressive motor neuron disease with autonomic and sensory disturbances, urine incontinence and sensory neuropathy. This patient was found to have V31A mutation in the SOD1 gene. Although slow progression has been previously observed in patients with ALS caused by several mutations in the SOD1 gene, symptoms unrelated with motor systems are very rare. In addition, MRI showed cerebellar and brainstem atrophy, a finding previously unreported in SOD1-related ALS. The COQ2 gene, a gene very recently reported to be associated with multiple system atrophy, as well as genes for spinocerebellar ataxias was analyzed, the result of which showed no mutation in this patient. The V31A mutation is thus likely to be associated with atypical ALS affecting multiple systems. DOI: 10.3109/21678421.2013.873051 PMID: 24611504 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35170113
1. Cell Prolif. 2022 Apr;55(4):e13202. doi: 10.1111/cpr.13202. Epub 2022 Feb 16. Enhancer RNA: What we know and what we can achieve. Han Z(1), Li W(1). Author information: (1)Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, China. Enhancers are important cis-acting elements that can regulate gene transcription and cell fate alongside promoters. In fact, many human cancers and diseases are associated with the malfunction of enhancers. Recent studies have shown that enhancers can produce enhancer RNAs (eRNAs) by RNA polymerase II. In this review, we discuss eRNA production, characteristics, functions and mechanics. eRNAs can determine chromatin accessibility, histone modification and gene expression by constructing a 'chromatin loop', thereby bringing enhancers to their target gene. eRNA can also be involved in the phase separation with enhancers and other proteins. eRNAs are abundant, and importantly, tissue-specific in tumours, various diseases and stem cells; thus, eRNAs can be a potential target for disease diagnosis and treatment. As eRNA is produced from the active transcription of enhancers and is involved in the regulation of cell fate, its manipulation will influence cell function, and therefore, it can be a new target for biological therapy. © 2022 The Authors. Cell Proliferation Published by John Wiley & Sons Ltd. DOI: 10.1111/cpr.13202 PMCID: PMC9055912 PMID: 35170113 [Indexed for MEDLINE] Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/31944157
1. Transcription. 2020 Feb;11(1):26-36. doi: 10.1080/21541264.2020.1713682. Epub 2020 Jan 16. Epigenetic plasticity of enhancers in cancer. Yao J(1), Chen J(1), Li LY(1), Wu M(1). Author information: (1)College of Life Sciences, Frontier Science Center for Immunology and Metabolism, Hubei Key Laboratory of Cell Homeostasis, Hubei Key Laboratory of Developmentally Originated Disease, Hubei Key Laboratory of Enteropathy, Wuhan University, Wuhan, Hubei, China. Enhancers are cis-acting elements with many sites bound by transcription factors and activate transcription over long distance. Histone modifications are critical for enhancer activity and utilized as hallmarks for the identification of putative enhancers. Monomethylation of histone H3 lysine 4 (H3K4me1) is the mark for enhancer priming; acetylation of histone H3 lysine 27 (H3K27ac) for active enhancers and trimethylation of histone H3 lysine 27 (H3K27me3) for silent enhancers. Recent studies from multiple groups have provided evidence that enhancer reprogramming, especially gain of enhancer activity, is closely related to tumorigenesis and cancer development. In this review, we will summarize the recent discoveries about enhancer regulation and the mechanisms of enhancer reprogramming in tumorigenesis, and discuss the potential application of enhancer manipulation in precision medicine. DOI: 10.1080/21541264.2020.1713682 PMCID: PMC7053964 PMID: 31944157 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/31993419
1. Front Cell Dev Biol. 2020 Jan 14;7:377. doi: 10.3389/fcell.2019.00377. eCollection 2019. Diversity and Emerging Roles of Enhancer RNA in Regulation of Gene Expression and Cell Fate. Arnold PR(1)(2), Wells AD(3), Li XC(2). Author information: (1)Texas A&M Health Science Center, College of Medicine, Bryan, TX, United States. (2)Immunobiology and Transplant Sciences, Department of Surgery, Houston Methodist Hospital, Houston, TX, United States. (3)Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States. Enhancers are cis-regulatory elements in the genome that cooperate with promoters to control target gene transcription. Unlike promoters, enhancers are not necessarily adjacent to target genes and can exert their functions regardless of enhancer orientations, positions and spatial segregations from target genes. Thus, for a long time, the question as to how enhancers act in a temporal and spatial manner attracted considerable attention. The recent discovery that enhancers are also abundantly transcribed raises interesting questions about the exact roles of enhancer RNA (eRNA) in gene regulation. In this review, we highlight the process of enhancer transcription and the diverse features of eRNA. We review eRNA functions, which include enhancer-promoter looping, chromatin modifying, and transcription regulating. As eRNA are transcribed from active enhancers, they exhibit tissue and lineage specificity, and serve as markers of cell state and function. Finally, we discuss the unique relationship between eRNA and super enhancers in phase separation wherein eRNA may contribute significantly to cell fate decisions. Copyright © 2020 Arnold, Wells and Li. DOI: 10.3389/fcell.2019.00377 PMCID: PMC6971116 PMID: 31993419
http://www.ncbi.nlm.nih.gov/pubmed/30026829
1. J Cancer. 2018 Jun 6;9(13):2334-2340. doi: 10.7150/jca.25829. eCollection 2018. Enhancer RNAs (eRNAs): New Insights into Gene Transcription and Disease Treatment. Ding M(1)(2), Liu Y(1)(3), Liao X(3), Zhan H(1)(3), Liu Y(3), Huang W(3). Author information: (1)Department of Urology, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Clinical Medicine College of Anhui Medical University, Shenzhen 518000, Guangdong, China. (2)Anhui Medical University, Hefei 230032, Anhui Province, China. (3)Department of Urology, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen 518000, China. Enhancers are cis-acting elements that have the ability to increase the expression of target genes. Recent studies have shown that enhancers can act as transcriptional units for the production of enhancer RNAs (eRNAs), which are hallmarks of activity enhancers and are involved in the regulation of gene transcription. The in-depth study of eRNAs is of great significance for us to better understand enhancer function and transcriptional regulation in various diseases. Therefore, eRNAs may be a potential therapeutic target for diseases. Here, we review the current knowledge of the characteristics of eRNAs, the molecular mechanisms of eRNAs action, as well as diseases related to dysregulation of eRNAs. DOI: 10.7150/jca.25829 PMCID: PMC6036709 PMID: 30026829 Conflict of interest statement: Competing Interests: The authors have declared that no competing interest exists.
http://www.ncbi.nlm.nih.gov/pubmed/29284524
1. Genome Biol. 2017 Dec 28;18(1):242. doi: 10.1186/s13059-017-1379-8. Bidirectional transcription initiation marks accessible chromatin and is not specific to enhancers. Young RS(1), Kumar Y(2), Bickmore WA(2), Taylor MS(3). Author information: (1)MRC Human Genetics Unit, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XU, UK. robert.young@igmm.ed.ac.uk. (2)MRC Human Genetics Unit, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XU, UK. (3)MRC Human Genetics Unit, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XU, UK. martin.taylor@igmm.ed.ac.uk. BACKGROUND: Enhancers are modular regulatory elements that are central to the spatial and temporal regulation of gene expression. Bidirectional transcription initiating at enhancers has been proposed to mark active enhancers and as such has been utilized to experimentally identify active enhancers de novo. RESULTS: Here, we show that bidirectional transcription initiation is a pervasive feature of accessible chromatin, including at enhancers, promoters, and other DNase hypersensitive regions not marked with canonical histone modification profiles. Transcription is less predictive for enhancer activity than epigenetic modifications such as H3K4me1 or the accessibility of DNA when measured both in enhancer assays and at endogenous loci. The stability of enhancer initiated transcripts does not influence measures of enhancer activity and we cannot detect evidence of purifying selection on the resulting enhancer RNAs within the human population. CONCLUSIONS: Our results indicate that bidirectional transcription initiation from accessible chromatin is not sufficient for, nor specific to, enhancer activity. Transcription initiating at enhancers may be a frequent by-product of promiscuous RNA polymerase initiation at accessible chromatin and is unlikely to generally play a functional role in enhancer activity. DOI: 10.1186/s13059-017-1379-8 PMCID: PMC5747114 PMID: 29284524 [Indexed for MEDLINE] Conflict of interest statement: ETHICS APPROVAL AND CONSENT TO PARTICIPATE: Not applicable. CONSENT FOR PUBLICATION: Not applicable. COMPETING INTERESTS: The authors declare that they have no competing interests. PUBLISHER’S NOTE: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
http://www.ncbi.nlm.nih.gov/pubmed/28239713
1. Mol Biosyst. 2017 Mar 28;13(4):767-774. doi: 10.1039/c7mb00054e. EnhancerPred2.0: predicting enhancers and their strength based on position-specific trinucleotide propensity and electron-ion interaction potential feature selection. He W(1), Jia C(1). Author information: (1)Department of Mathematics, Dalian Maritime University, No. 1 Linghai Road, Dalian 116026, China. cangzhijia@dlmu.edu.cn. Enhancers are cis-acting elements that play major roles in upregulating eukaryotic gene expression by providing binding sites for transcription factors and their complexes. Because enhancers are highly cell/tissue specific, lack common motifs, and are far from the target gene, the systematic and precise identification of enhancer regions in DNA sequences is a big challenge. In this study, we developed an enhancer prediction method called EnhancerPred2.0 by combining position-specific trinucleotide propensity (PSTNP) information with the electron-ion interaction potential (EIIP) values for trinucleotides, to predict enhancers and their subgroups. To obtain the optimal combination of features, F-score values were used in a two-step wrapper-based feature selection method, which was applied in a high dimensional feature vector from PSTNP and EIIP. Finally, 126 optimized features from PSTNP combined with 32 optimized features from EIIP yielded the best performance for identifying enhancers and non-enhancers, with an overall accuracy (Acc) of 88.27% and a Matthews correlation coefficient (MCC) of 0.77. Additionally, 198 features from PSTNP combined with 37 features from EIIP yielded the best performance for identifying strong and weak enhancers, with an overall Acc of 98.05% and a MCC of 0.96. Rigorous jackknife tests indicated that EnhancerPred2.0 was significantly better than the existing enhancer prediction methods in both overall accuracy and stability. DOI: 10.1039/c7mb00054e PMID: 28239713 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/30139328
1. BMC Genomics. 2018 Aug 23;19(1):633. doi: 10.1186/s12864-018-5016-z. Nascent RNA sequencing analysis provides insights into enhancer-mediated gene regulation. Wang J(1)(2), Zhao Y(3), Zhou X(4), Hiebert SW(3)(5), Liu Q(6)(7), Shyr Y(8)(9). Author information: (1)Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA. (2)Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA. (3)Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA. (4)Department of Biological Science, Vanderbilt University, Nashville, TN, USA. (5)Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA. (6)Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA. qi.liu@vanderbilt.edu. (7)Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA. qi.liu@vanderbilt.edu. (8)Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA. yu.shyr@vanderbilt.edu. (9)Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA. yu.shyr@vanderbilt.edu. BACKGROUND: Enhancers are distal cis-regulatory elements that control gene expression. Despite an increasing appreciation of the importance of enhancers in cellular function and disease, our knowledge of enhancer-regulated transcription is very limited. Nascent RNA sequencing technologies, such as global nuclear run-on sequencing (GRO-seq) and precision run-on sequencing (PRO-seq), not only provide a direct and reliable measurement of enhancer activity, but also allow for quantifying transcription of enhancers and target genes simultaneously, making these technologies extremely useful for exploring enhancer-mediated regulation. RESULTS: Nascent RNA sequencing analysis (NRSA) provides a comprehensive view of enhancer-mediated gene regulation. NRSA not only outperforms existing methods for enhancer identification, but also enables annotation and quantification of active enhancers, and prediction of their target genes. Furthermore, NRSA identifies functionally important enhancers by integrating 1) nascent transcriptional changes in enhancers and their target genes and 2) binding profiles from regulator(s) of interest. Applied to wildtype and histone deacetylase 3 (Hdac3) knockout mouse livers, NRSA showed that HDAC3 regulates RNA polymerase recruitment through both proximal (promoter) and distal (enhancer) regulatory elements. Integrating ChIP-seq with PRO-seq data, NRSA prioritized enhancers based on their potential contribution to mediating HDAC3 regulation. CONCLUSIONS: NRSA will greatly facilitate the usage of nascent RNA sequencing techniques and accelerate the study of enhancer-mediated regulation. DOI: 10.1186/s12864-018-5016-z PMCID: PMC6107967 PMID: 30139328 [Indexed for MEDLINE] Conflict of interest statement: ETHICS APPROVAL AND CONSENT TO PARTICIPATE: The genetically engineered mice were created, bred and housed under specific pathogen-free conditions at and in accordance with guidelines set forth by Vanderbilt University Medical Center. Animal ethics was approved by the Vanderbilt University Institutional Animal Care and Use Committee (IACUC), protocol number M/12/021. CONSENT FOR PUBLICATION: Not applicable. COMPETING INTERESTS: The authors declare that they have no competing interests. PUBLISHER’S NOTE: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
http://www.ncbi.nlm.nih.gov/pubmed/28334114
1. Bioinformatics. 2017 Jul 1;33(13):1930-1936. doi: 10.1093/bioinformatics/btx105. BiRen: predicting enhancers with a deep-learning-based model using the DNA sequence alone. Yang B(1), Liu F(1)(2), Ren C(1), Ouyang Z(1), Xie Z(3), Bo X(1), Shu W(1). Author information: (1)Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850. (2)Department of Information, The 188th Hospital of Chaozhou, Chaozhou 521000. (3)Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China. MOTIVATION: Enhancer elements are noncoding stretches of DNA that play key roles in controlling gene expression programmes. Despite major efforts to develop accurate enhancer prediction methods, identifying enhancer sequences continues to be a challenge in the annotation of mammalian genomes. One of the major issues is the lack of large, sufficiently comprehensive and experimentally validated enhancers for humans or other species. Thus, the development of computational methods based on limited experimentally validated enhancers and deciphering the transcriptional regulatory code encoded in the enhancer sequences is urgent. RESULTS: We present a deep-learning-based hybrid architecture, BiRen, which predicts enhancers using the DNA sequence alone. Our results demonstrate that BiRen can learn common enhancer patterns directly from the DNA sequence and exhibits superior accuracy, robustness and generalizability in enhancer prediction relative to other state-of-the-art enhancer predictors based on sequence characteristics. Our BiRen will enable researchers to acquire a deeper understanding of the regulatory code of enhancer sequences. AVAILABILITY AND IMPLEMENTATION: Our BiRen method can be freely accessed at https://github.com/wenjiegroup/BiRen . CONTACT: shuwj@bmi.ac.cn or boxc@bmi.ac.cn. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online. © The Author 2017. Published by Oxford University Press. All rights reserved. For Permissions, please e-mail: journals.permissions@oup.com DOI: 10.1093/bioinformatics/btx105 PMID: 28334114 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/28716036
1. BMC Genomics. 2017 Jul 17;18(1):536. doi: 10.1186/s12864-017-3934-9. Short DNA sequence patterns accurately identify broadly active human enhancers. Colbran LL(1), Chen L(2), Capra JA(3)(4)(5). Author information: (1)Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, 37235, USA. (2)Department of Biological Sciences, Vanderbilt University, Nashville, TN, 37235, USA. (3)Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, 37235, USA. tony.capra@vanderbilt.edu. (4)Department of Biological Sciences, Vanderbilt University, Nashville, TN, 37235, USA. tony.capra@vanderbilt.edu. (5)Center for Structural Biology, Departments of Biomedical Informatics and Computer Science, Vanderbilt University, Nashville, TN, 37235, USA. tony.capra@vanderbilt.edu. BACKGROUND: Enhancers are DNA regulatory elements that influence gene expression. There is substantial diversity in enhancers' activity patterns: some enhancers drive expression in a single cellular context, while others are active across many. Sequence characteristics, such as transcription factor (TF) binding motifs, influence the activity patterns of regulatory sequences; however, the regulatory logic through which specific sequences drive enhancer activity patterns is poorly understood. Recent analysis of Drosophila enhancers suggested that short dinucleotide repeat motifs (DRMs) are general enhancer sequence features that drive broad regulatory activity. However, it is not known whether the regulatory role of DRMs is conserved across species. RESULTS: We performed a comprehensive analysis of the relationship between short DNA sequence patterns, including DRMs, and human enhancer activity in 38,538 enhancers across 411 different contexts. In a machine-learning framework, the occurrence patterns of short sequence motifs accurately predicted broadly active human enhancers. However, DRMs alone were weakly predictive of broad enhancer activity in humans and showed different enrichment patterns than in Drosophila. In general, GC-rich sequence motifs were significantly associated with broad enhancer activity, and consistent with this enrichment, broadly active human TFs recognize GC-rich motifs. CONCLUSIONS: Our results reveal the importance of specific sequence motifs in broadly active human enhancers, demonstrate the lack of evolutionary conservation of the role of DRMs, and provide a computational framework for investigating the logic of enhancer sequences. DOI: 10.1186/s12864-017-3934-9 PMCID: PMC5512948 PMID: 28716036 [Indexed for MEDLINE] Conflict of interest statement: ETHICS APPROVAL AND CONSENT TO PARTICIPATE: Not applicable. CONSENT FOR PUBLICATION: Not applicable. COMPETING INTERESTS: The authors declare they have no competing interests. PUBLISHER’S NOTE: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
http://www.ncbi.nlm.nih.gov/pubmed/33320871
1. PLoS One. 2020 Dec 15;15(12):e0243791. doi: 10.1371/journal.pone.0243791. eCollection 2020. PEREGRINE: A genome-wide prediction of enhancer to gene relationships supported by experimental evidence. Mills C(1), Muruganujan A(2), Ebert D(2), Marconett CN(3)(4)(5), Lewinger JP(1), Thomas PD(2), Mi H(2). Author information: (1)Division of Biostatistics, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America. (2)Division of Bioinformatics, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America. (3)Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America. (4)Department of Biochemistry and Molecular Medicine, Keck School of Medicine USC, Los Angeles, CA, United States of America. (5)Norris Cancer Center, Keck School of Medicine USC, Los Angeles, CA, United States of America. Enhancers are powerful and versatile agents of cell-type specific gene regulation, which are thought to play key roles in human disease. Enhancers are short DNA elements that function primarily as clusters of transcription factor binding sites that are spatially coordinated to regulate expression of one or more specific target genes. These regulatory connections between enhancers and target genes can therefore be characterized as enhancer-gene links that can affect development, disease, and homeostatic cellular processes. Despite their implication in disease and the establishment of cell identity during development, most enhancer-gene links remain unknown. Here we introduce a new, publicly accessible database of predicted enhancer-gene links, PEREGRINE. The PEREGRINE human enhancer-gene links interactive web interface incorporates publicly available experimental data from ChIA-PET, eQTL, and Hi-C assays across 78 cell and tissue types to link 449,627 enhancers to 17,643 protein-coding genes. These enhancer-gene links are made available through the new Enhancer module of the PANTHER database and website where the user may easily access the evidence for each enhancer-gene link, as well as query by target gene and enhancer location. DOI: 10.1371/journal.pone.0243791 PMCID: PMC7737992 PMID: 33320871 [Indexed for MEDLINE] Conflict of interest statement: The authors have declared that no competing interests exist.
http://www.ncbi.nlm.nih.gov/pubmed/31665430
1. Nucleic Acids Res. 2020 Jan 8;48(D1):D51-D57. doi: 10.1093/nar/gkz973. ENdb: a manually curated database of experimentally supported enhancers for human and mouse. Bai X(1), Shi S(1), Ai B(1), Jiang Y(1), Liu Y(1), Han X(1), Xu M(1), Pan Q(1), Wang F(1), Wang Q(1), Zhang J(1), Li X(1), Feng C(1), Li Y(1), Wang Y(1), Song Y(1), Feng K(1), Li C(1). Author information: (1)School of Medical Informatics, Daqing Campus, Harbin Medical University. Daqing 163319, China. Enhancers are a class of cis-regulatory elements that can increase gene transcription by forming loops in intergenic regions, introns and exons. Enhancers, as well as their associated target genes, and transcription factors (TFs) that bind to them, are highly associated with human disease and biological processes. Although some enhancer databases have been published, most only focus on enhancers identified by high-throughput experimental techniques. Therefore, it is highly desirable to construct a comprehensive resource of manually curated enhancers and their related information based on low-throughput experimental evidences. Here, we established a comprehensive manually-curated enhancer database for human and mouse, which provides a resource for experimentally supported enhancers, and to annotate the detailed information of enhancers. The current release of ENdb documents 737 experimentally validated enhancers and their related information, including 384 target genes, 263 TFs, 110 diseases and 153 functions in human and mouse. Moreover, the enhancer-related information was supported by experimental evidences, such as RNAi, in vitro knockdown, western blotting, qRT-PCR, luciferase reporter assay, chromatin conformation capture (3C) and chromosome conformation capture-on-chip (4C) assays. ENdb provides a user-friendly interface to query, browse and visualize the detailed information of enhancers. The database is available at http://www.licpathway.net/ENdb. © The Author(s) 2019. Published by Oxford University Press on behalf of Nucleic Acids Research. DOI: 10.1093/nar/gkz973 PMCID: PMC7145688 PMID: 31665430 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/27662874
1. Methods Mol Biol. 2017;1468:121-38. doi: 10.1007/978-1-4939-4035-6_10. Computational Approaches for Mining GRO-Seq Data to Identify and Characterize Active Enhancers. Nagari A(1)(2), Murakami S(1)(2)(3), Malladi VS(1)(2), Kraus WL(4)(5)(6). Author information: (1)The Laboratory of Signaling and Gene Expression, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-8511, USA. (2)The Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, 75390-8511, USA. (3)Program in Genetics, Development and Disease, Graduate School of Biomedical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA. (4)The Laboratory of Signaling and Gene Expression, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-8511, USA. LEE.KRAUS@utsouthwestern.edu. (5)The Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, 75390-8511, USA. LEE.KRAUS@utsouthwestern.edu. (6)Program in Genetics, Development and Disease, Graduate School of Biomedical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA. LEE.KRAUS@utsouthwestern.edu. Transcriptional enhancers are DNA regulatory elements that are bound by transcription factors and act to positively regulate the expression of nearby or distally located target genes. Enhancers have many features that have been discovered using genomic analyses. Recent studies have shown that active enhancers recruit RNA polymerase II (Pol II) and are transcribed, producing enhancer RNAs (eRNAs). GRO-seq, a method for identifying the location and orientation of all actively transcribing RNA polymerases across the genome, is a powerful approach for monitoring nascent enhancer transcription. Furthermore, the unique pattern of enhancer transcription can be used to identify enhancers in the absence of any information about the underlying transcription factors. Here, we describe the computational approaches required to identify and analyze active enhancers using GRO-seq data, including data pre-processing, alignment, and transcript calling. In addition, we describe protocols and computational pipelines for mining GRO-seq data to identify active enhancers, as well as known transcription factor binding sites that are transcribed. Furthermore, we discuss approaches for integrating GRO-seq-based enhancer data with other genomic data, including target gene expression and function. Finally, we describe molecular biology assays that can be used to confirm and explore further the function of enhancers that have been identified using genomic assays. Together, these approaches should allow the user to identify and explore the features and biological functions of new cell type-specific enhancers. DOI: 10.1007/978-1-4939-4035-6_10 PMCID: PMC5522910 PMID: 27662874 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/29583027
1. Epigenomics. 2018 Apr 1;10(4):483-498. doi: 10.2217/epi-2017-0157. Epub 2018 Mar 27. Enhancer talk. Snetkova V(1)(2), Skok JA(1). Author information: (1)Department of Pathology, New York University School of Medicine, 550 First Avenue, MSB 599, New York, NY 10016, USA. (2)MS 84-171, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA. Enhancers are short noncoding segments of DNA (100-1000 bp) that control the temporal and spatial activity of genes in an orientation-independent manner. They can be separated from their target genes by large distances and are thus known as distal regulatory elements. One consequence of the variability in the distance separating enhancers and their target promoters is that it is difficult to determine which elements are involved in the regulation of a particular gene. Moreover, enhancers can be found in clusters in which multiple regulatory elements control expression of the same target gene. However, little is known about how the individual elements contribute to gene expression. Here, we describe how chromatin conformation promotes and constraints enhancer activity. Further, we discuss enhancer clusters and what is known about the contribution of individual elements to the regulation of target genes. Finally, we examine the reliability of different methods used to identify enhancers. DOI: 10.2217/epi-2017-0157 PMCID: PMC5925435 PMID: 29583027 [Indexed for MEDLINE] Conflict of interest statement: Financial & competing interests disclosure JA Skok is supported by NIH grant R35GM122515, 4P30CA016087-36 Cancer Center Support Grant NIH/NCI (Neel) and 2R01CA140729-06A1 NIH/NCI (Carroll). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed. No writing assistance was utilized in the production of this manuscript.
http://www.ncbi.nlm.nih.gov/pubmed/32197056
1. Annu Rev Biochem. 2020 Jun 20;89:213-234. doi: 10.1146/annurev-biochem-011420-095916. Epub 2020 Mar 20. Evaluating Enhancer Function and Transcription. Field A(1), Adelman K(1). Author information: (1)Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA; email: karen_adelman@hms.harvard.edu. Cell-type- and condition-specific profiles of gene expression require coordination between protein-coding gene promoters and cis-regulatory sequences called enhancers. Enhancers can stimulate gene activity at great genomic distances from their targets, raising questions about how enhancers communicate with specific gene promoters and what molecular mechanisms underlie enhancer function. Characterization of enhancer loci has identified the molecular features of active enhancers that accompany the binding of transcription factors and local opening of chromatin. These characteristics include coactivator recruitment, histone modifications, and noncoding RNA transcription. However, it remains unclear which of these features functionally contribute to enhancer activity. Here, we discuss what is known about how enhancers regulate their target genes and how enhancers and promoters communicate. Further, we describe recent data demonstrating many similarities between enhancers and the gene promoters they control, and we highlight unanswered questions in the field, such as the potential roles of transcription at enhancers. DOI: 10.1146/annurev-biochem-011420-095916 PMID: 32197056 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/33431820
1. Nat Commun. 2021 Jan 11;12(1):223. doi: 10.1038/s41467-020-20400-z. BET inhibition disrupts transcription but retains enhancer-promoter contact. Crump NT(1), Ballabio E(1), Godfrey L(1), Thorne R(1), Repapi E(2), Kerry J(1), Tapia M(1)(3)(4), Hua P(5), Lagerholm C(6), Filippakopoulos P(7), Davies JOJ(5), Milne TA(8). Author information: (1)MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK. (2)MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK. (3)The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark. (4)Biotech Research and Innovation Centre (BRIC), Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark. (5)MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK. (6)Wolfson Imaging Centre Oxford, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK. (7)Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, UK. (8)MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK. thomas.milne@imm.ox.ac.uk. Enhancers are DNA sequences that enable complex temporal and tissue-specific regulation of genes in higher eukaryotes. Although it is not entirely clear how enhancer-promoter interactions can increase gene expression, this proximity has been observed in multiple systems at multiple loci and is thought to be essential for the maintenance of gene expression. Bromodomain and Extra-Terminal domain (BET) and Mediator proteins have been shown capable of forming phase condensates and are thought to be essential for super-enhancer function. Here, we show that targeting of cells with inhibitors of BET proteins or pharmacological degradation of BET protein Bromodomain-containing protein 4 (BRD4) has a strong impact on transcription but very little impact on enhancer-promoter interactions. Dissolving phase condensates reduces BRD4 and Mediator binding at enhancers and can also strongly affect gene transcription, without disrupting enhancer-promoter interactions. These results suggest that activation of transcription and maintenance of enhancer-promoter interactions are separable events. Our findings further indicate that enhancer-promoter interactions are not dependent on high levels of BRD4 and Mediator, and are likely maintained by a complex set of factors including additional activator complexes and, at some sites, CTCF and cohesin. DOI: 10.1038/s41467-020-20400-z PMCID: PMC7801379 PMID: 33431820 [Indexed for MEDLINE] Conflict of interest statement: T.A.M. is a founding shareholder of OxStem Oncology (OSO), a subsidiary company of OxStem Ltd. J.O.J.D. is a co-founder of Nucleome Therapeutics Ltd. to which he provides consultancy. All other authors have no competing interests.
http://www.ncbi.nlm.nih.gov/pubmed/21737276
1. Curr Biol. 2011 Jul 26;21(14):1186-96. doi: 10.1016/j.cub.2011.05.056. Epub 2011 Jul 7. Rapid evolutionary rewiring of a structurally constrained eye enhancer. Swanson CI(1), Schwimmer DB, Barolo S. Author information: (1)Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA. Comment in Curr Biol. 2011 Jul 26;21(14):R542-3. doi: 10.1016/j.cub.2011.06.026. BACKGROUND: Enhancers are genomic cis-regulatory sequences that integrate spatiotemporal signals to control gene expression. Enhancer activity depends on the combination of bound transcription factors as well as-in some cases-the arrangement and spacing of binding sites for these factors. Here, we examine evolutionary changes to the sequence and structure of sparkling, a Notch/EGFR/Runx-regulated enhancer that activates the dPax2 gene in cone cells of the developing Drosophila eye. RESULTS: Despite functional and structural constraints on its sequence, sparkling has undergone major reorganization in its recent evolutionary history. Our data suggest that the relative strengths of the various regulatory inputs into sparkling change rapidly over evolutionary time, such that reduced input from some factors is compensated by increased input from different regulators. These gains and losses are at least partly responsible for the changes in enhancer structure that we observe. Furthermore, stereotypical spatial relationships between certain binding sites ("grammar elements") can be identified in all sparkling orthologs-although the sites themselves are often recently derived. We also find that low binding affinity for the Notch-regulated transcription factor Su(H), a conserved property of sparkling, is required to prevent ectopic responses to Notch in noncone cells. CONCLUSIONS: Rapid DNA sequence turnover does not imply either the absence of critical cis-regulatory information or the absence of structural rules. Our findings demonstrate that even a severely constrained cis-regulatory sequence can be significantly rewired over a short evolutionary timescale. Copyright © 2011 Elsevier Ltd. All rights reserved. DOI: 10.1016/j.cub.2011.05.056 PMCID: PMC3143281 PMID: 21737276 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/16002789
1. Nucleic Acids Res. 2005 Jul 7;33(12):3743-50. doi: 10.1093/nar/gki689. Print 2005. DNA looping induced by a transcriptional enhancer in vivo. Petrascheck M(1), Escher D, Mahmoudi T, Verrijzer CP, Schaffner W, Barberis A. Author information: (1)Institute of Molecular Biology, University of Zurich Winterthurerstrasse 190, CH-8057 Zurich, Switzerland. mpetrasc@fhcrc.org Enhancers are DNA sequences that can activate gene transcription from remote positions. In yeast, regulatory sequences that are functionally equivalent to the metazoan enhancers are called upstream activating sequences (UASs). UASs show a lower degree of flexibility than their metazoan counterparts, but can nevertheless activate transcription from a distance of >1000 bp from the promoter. One of several models for the mechanism of action of transcriptional enhancers proposes that enhancer-bound activating proteins contact promoter-bound transcription factors and thereby get in close proximity to the promoter region with concomitant looping of the intervening DNA. We tested the mode of enhancer activity in yeast. A polymerase II-transcribed gene was paired with a remote, inducible enhancer. An independent reporter system was inserted next to the promoter to monitor the potential modes of enhancer activity. Our results show that the enhancer activated the reporter system only in the presence of a functional promoter. We also demonstrate that the heterologous expression of GAGA, a factor known to facilitate DNA loop formation, allows enhancer action in yeast over a distance of 3000 bp. DOI: 10.1093/nar/gki689 PMCID: PMC1174898 PMID: 16002789 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/30758829
1. Methods Mol Biol. 2019;1935:203-221. doi: 10.1007/978-1-4939-9057-3_14. Experimental and Computational Approaches for Single-Cell Enhancer Perturbation Assay. Xie S(1), Hon GC(2). Author information: (1)Department of Obstetrics and Gynecology, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA. (2)Department of Obstetrics and Gynecology, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA. Gary.Hon@UTSouthwestern.edu. Transcriptional enhancers drive cell-type-specific gene expression patterns, and thus play key roles in development and disease. Large-scale consortia have extensively cataloged >one million putative enhancers encoded in the human genome. But few enhancers have been endogenously tested for function. For almost all enhancers, it remains unknown what genes they target and how much they contribute to target gene expression. We have previously developed a method called Mosaic-seq, which enables the high-throughput interrogation of enhancer activity by performing pooled CRISPRi-based epigenetic suppression of enhancers with a single-cell transcriptomic readout. Here, we describe an optimized version of this method, Mosaic-seq2. We have made several key improvements that have significantly simplified the library preparation process and increased the overall sensitivity and throughput of the method. DOI: 10.1007/978-1-4939-9057-3_14 PMID: 30758829 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/32664620
1. Cells. 2020 Jul 12;9(7):1675. doi: 10.3390/cells9071675. Evolution of Regulated Transcription. Bylino OV(1), Ibragimov AN(1)(2), Shidlovskii YV(1)(3). Author information: (1)Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia. (2)Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia. (3)I.M. Sechenov First Moscow State Medical University, 8, bldg. 2 Trubetskaya St., 119048 Moscow, Russia. The genomes of all organisms abound with various cis-regulatory elements, which control gene activity. Transcriptional enhancers are a key group of such elements in eukaryotes and are DNA regions that form physical contacts with gene promoters and precisely orchestrate gene expression programs. Here, we follow gradual evolution of this regulatory system and discuss its features in different organisms. In eubacteria, an enhancer-like element is often a single regulatory element, is usually proximal to the core promoter, and is occupied by one or a few activators. Activation of gene expression in archaea is accompanied by the recruitment of an activator to several enhancer-like sites in the upstream promoter region. In eukaryotes, activation of expression is accompanied by the recruitment of activators to multiple enhancers, which may be distant from the core promoter, and the activators act through coactivators. The role of the general DNA architecture in transcription control increases in evolution. As a whole, it can be seen that enhancers of multicellular eukaryotes evolved from the corresponding prototypic enhancer-like regulatory elements with the gradually increasing genome size of organisms. DOI: 10.3390/cells9071675 PMCID: PMC7408454 PMID: 32664620 [Indexed for MEDLINE] Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/23124110
1. Biochim Biophys Acta. 2012 Nov-Dec;1819(11-12):1217-27. doi: 10.1016/j.bbagrm.2012.10.008. Epub 2012 Nov 1. Long distance relationships: enhancer-promoter communication and dynamic gene transcription. Marsman J(1), Horsfield JA. Author information: (1)Department of Pathology, The University of Otago, Dunedin, New Zealand. The three-dimensional regulation of gene transcription involves loop formation between enhancer and promoter elements, controlling spatiotemporal gene expression in multicellular organisms. Enhancers are usually located in non-coding DNA and can activate gene transcription by recruiting transcription factors, chromatin remodeling factors and RNA Polymerase II. Research over the last few years has revealed that enhancers have tell-tale characteristics that facilitate their detection by several approaches, although the hallmarks of enhancers are not always uniform. Enhancers likely play an important role in the activation of genes by functioning as a primary point of contact for transcriptional activators, and by making physical contact with gene promoters often by means of a chromatin loop. Although numerous transcriptional regulators participate in the formation of chromatin loops that bring enhancers into proximity with promoters, the mechanism(s) of enhancer-promoter connectivity remain enigmatic. Here we discuss enhancer function, review some of the many proteins shown to be involved in establishing enhancer-promoter loops, and describe the dynamics of enhancer-promoter contacts during development, differentiation and in specific cell types. Copyright © 2012 Elsevier B.V. All rights reserved. DOI: 10.1016/j.bbagrm.2012.10.008 PMID: 23124110 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/27325706
1. Cold Spring Harb Symp Quant Biol. 2015;80:27-32. doi: 10.1101/sqb.2015.80.027227. Regulatory Principles Governing Tissue Specificity of Developmental Enhancers. Farley EK(1), Olson KM(1), Levine MS(1). Author information: (1)Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey 08544. Transcriptional enhancers are short segments of genomic DNA (50 bp to 1 kb in length) that can work over long distances (≥1 Mb) to regulate gene expression in specific cells and tissues. Genomic assays have identified on the order of 400,000 to one million putative enhancers in the human genome (e.g., ENCODE Consortium). This suggests that a typical gene is regulated by tens of enhancers, ensuring stringent regulation of gene expression in response to a variety of intrinsic and external signals. Despite the discovery of the first transcriptional enhancer more than 30 years ago, we know surprisingly little about how enhancers regulate gene expression. In particular, the relationship between primary DNA sequence and enhancer specificity remains obscure. Here we summarize recent high-throughput studies in whole embryos aimed at the systematic identification of the sequence and organizational constraints underlying enhancer function and specificity. Copyright © 2015 Cold Spring Harbor Laboratory Press; all rights reserved. DOI: 10.1101/sqb.2015.80.027227 PMCID: PMC4970742 PMID: 27325706 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/28760812
1. Development. 2017 Sep 1;144(17):3095-3101. doi: 10.1242/dev.149427. Epub 2017 Jul 31. Functional regulatory evolution outside of the minimal even-skipped stripe 2 enhancer. Crocker J(1), Stern DL(2). Author information: (1)Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, VA 20147, USA justin.crocker@embl.de. (2)Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, VA 20147, USA. Transcriptional enhancers are regions of DNA that drive precise patterns of gene expression. Although many studies have elucidated how individual enhancers can evolve, most of this work has focused on what are called 'minimal' enhancers, the smallest DNA regions that drive expression that approximates an aspect of native gene expression. Here, we explore how the Drosophila erecta even-skipped (eve) locus has evolved by testing its activity in the divergent D. melanogaster genome. We found, as has been reported previously, that the D. erecta eve stripe 2 enhancer (eveS2) fails to drive appreciable expression in D. melanogaster However, we found that a large transgene carrying the entire D. erecta eve locus drives normal eve expression, including in stripe 2. We performed a functional dissection of the region upstream of the D. erecta eveS2 region and found multiple Zelda motifs that are required for normal expression. Our results illustrate how sequences outside of minimal enhancer regions can evolve functionally through mechanisms other than changes in transcription factor-binding sites that drive patterning. © 2017. Published by The Company of Biologists Ltd. DOI: 10.1242/dev.149427 PMID: 28760812 [Indexed for MEDLINE] Conflict of interest statement: Competing interestsThe authors declare no competing or financial interests.
http://www.ncbi.nlm.nih.gov/pubmed/25715743
1. Cell Mol Life Sci. 2015 Jun;72(12):2361-75. doi: 10.1007/s00018-015-1871-9. Epub 2015 Feb 26. Eukaryotic enhancers: common features, regulation, and participation in diseases. Erokhin M(1), Vassetzky Y, Georgiev P, Chetverina D. Author information: (1)Department of the Control of Genetic Processes, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov St., Moscow, 119334, Russia. Enhancers are positive DNA regulatory sequences controlling temporal and tissue-specific gene expression. These elements act independently of their orientation and distance relative to the promoters of target genes. Enhancers act through a variety of transcription factors that ensure their correct match with target promoters and consequent gene activation. There is a growing body of evidence on association of enhancers with transcription factors, co-activators, histone chromatin marks, and lncRNAs. Alterations in enhancers lead to misregulation of gene expression, causing a number of human diseases. In this review, we focus on the common characteristics of enhancers required for transcription stimulation. DOI: 10.1007/s00018-015-1871-9 PMCID: PMC11114076 PMID: 25715743 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/32737473
1. Nat Methods. 2020 Aug;17(8):807-814. doi: 10.1038/s41592-020-0907-8. Epub 2020 Jul 29. Supervised enhancer prediction with epigenetic pattern recognition and targeted validation. Sethi A(#)(1), Gu M(#)(2)(3), Gumusgoz E(4), Chan L(5), Yan KK(1), Rozowsky J(1), Barozzi I(6), Afzal V(6), Akiyama JA(6), Plajzer-Frick I(6), Yan C(1), Novak CS(6), Kato M(6), Garvin TH(6), Pham Q(6), Harrington A(6), Mannion BJ(6), Lee EA(6), Fukuda-Yuzawa Y(6), Visel A(6), Dickel DE(6), Yip KY(7), Sutton R(4), Pennacchio LA(6), Gerstein M(8)(9)(10). Author information: (1)Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA. (2)Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA. (3)Department of Computer Science, Yale University, New Haven, CT, USA. (4)Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA. (5)School of Medicine, The Chinese University of Hong Kong, Hong Kong, China. (6)Functional Genomics Department, Lawrence Berkeley National Laboratory, Berkeley, CA, USA. (7)Department of Computer Science, The Chinese University of Hong Kong, Hong Kong, China. (8)Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA. mark@gersteinlab.org. (9)Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA. mark@gersteinlab.org. (10)Department of Computer Science, Yale University, New Haven, CT, USA. mark@gersteinlab.org. (#)Contributed equally Enhancers are important non-coding elements, but they have traditionally been hard to characterize experimentally. The development of massively parallel assays allows the characterization of large numbers of enhancers for the first time. Here, we developed a framework using Drosophila STARR-seq to create shape-matching filters based on meta-profiles of epigenetic features. We integrated these features with supervised machine-learning algorithms to predict enhancers. We further demonstrated that our model could be transferred to predict enhancers in mammals. We comprehensively validated the predictions using a combination of in vivo and in vitro approaches, involving transgenic assays in mice and transduction-based reporter assays in human cell lines (153 enhancers in total). The results confirmed that our model can accurately predict enhancers in different species without re-parameterization. Finally, we examined the transcription factor binding patterns at predicted enhancers versus promoters. We demonstrated that these patterns enable the construction of a secondary model that effectively distinguishes enhancers and promoters. DOI: 10.1038/s41592-020-0907-8 PMCID: PMC8073243 PMID: 32737473 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34879086
1. PLoS One. 2021 Dec 8;16(12):e0260432. doi: 10.1371/journal.pone.0260432. eCollection 2021. A network-based method for predicting disease-associated enhancers. Le DH(1). Author information: (1)School of Computer Science and Engineering, Thuyloi University, Hanoi, Vietnam. BACKGROUND: Enhancers regulate transcription of target genes, causing a change in expression level. Thus, the aberrant activity of enhancers can lead to diseases. To date, a large number of enhancers have been identified, yet a small portion of them have been found to be associated with diseases. This raises a pressing need to develop computational methods to predict associations between diseases and enhancers. RESULTS: In this study, we assumed that enhancers sharing target genes could be associated with similar diseases to predict the association. Thus, we built an enhancer functional interaction network by connecting enhancers significantly sharing target genes, then developed a network diffusion method RWDisEnh, based on a random walk with restart algorithm, on networks of diseases and enhancers to globally measure the degree of the association between diseases and enhancers. RWDisEnh performed best when the disease similarities are integrated with the enhancer functional interaction network by known disease-enhancer associations in the form of a heterogeneous network of diseases and enhancers. It was also superior to another network diffusion method, i.e., PageRank with Priors, and a neighborhood-based one, i.e., MaxLink, which simply chooses the closest neighbors of known disease-associated enhancers. Finally, we showed that RWDisEnh could predict novel enhancers, which are either directly or indirectly associated with diseases. CONCLUSIONS: Taken together, RWDisEnh could be a potential method for predicting disease-enhancer associations. DOI: 10.1371/journal.pone.0260432 PMCID: PMC8654176 PMID: 34879086 [Indexed for MEDLINE] Conflict of interest statement: The authors have declared that no competing interests exist.
http://www.ncbi.nlm.nih.gov/pubmed/27895109
1. Genome Res. 2017 Feb;27(2):246-258. doi: 10.1101/gr.210930.116. Epub 2016 Nov 28. Enhancers and super-enhancers have an equivalent regulatory role in embryonic stem cells through regulation of single or multiple genes. Moorthy SD(1), Davidson S(1), Shchuka VM(1), Singh G(1), Malek-Gilani N(1), Langroudi L(1), Martchenko A(1), So V(1), Macpherson NN(1), Mitchell JA(1)(2). Author information: (1)Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada. (2)Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, Ontario M5S 3G5, Canada. Transcriptional enhancers are critical for maintaining cell-type-specific gene expression and driving cell fate changes during development. Highly transcribed genes are often associated with a cluster of individual enhancers such as those found in locus control regions. Recently, these have been termed stretch enhancers or super-enhancers, which have been predicted to regulate critical cell identity genes. We employed a CRISPR/Cas9-mediated deletion approach to study the function of several enhancer clusters (ECs) and isolated enhancers in mouse embryonic stem (ES) cells. Our results reveal that the effect of deleting ECs, also classified as ES cell super-enhancers, is highly variable, resulting in target gene expression reductions ranging from 12% to as much as 92%. Partial deletions of these ECs which removed only one enhancer or a subcluster of enhancers revealed partially redundant control of the regulated gene by multiple enhancers within the larger cluster. Many highly transcribed genes in ES cells are not associated with a super-enhancer; furthermore, super-enhancer predictions ignore 81% of the potentially active regulatory elements predicted by cobinding of five or more pluripotency-associated transcription factors. Deletion of these additional enhancer regions revealed their robust regulatory role in gene transcription. In addition, select super-enhancers and enhancers were identified that regulated clusters of paralogous genes. We conclude that, whereas robust transcriptional output can be achieved by an isolated enhancer, clusters of enhancers acting on a common target gene act in a partially redundant manner to fine tune transcriptional output of their target genes. © 2017 Moorthy et al.; Published by Cold Spring Harbor Laboratory Press. DOI: 10.1101/gr.210930.116 PMCID: PMC5287230 PMID: 27895109 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35552697
1. Genome Biol Evol. 2022 May 31;14(6):evac071. doi: 10.1093/gbe/evac071. Pleiotropic Enhancers are Ubiquitous Regulatory Elements in the Human Genome. Laiker I(1), Frankel N(1)(2). Author information: (1)Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Universidad de Buenos Aires (UBA), Buenos Aires 1428, Argentina. (2)Departamento de Ecología, Genética y Evolución, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), Buenos Aires 1428, Argentina. Enhancers are regulatory elements of genomes that determine spatio-temporal patterns of gene expression. The human genome contains a vast number of enhancers, which largely outnumber protein-coding genes. Historically, enhancers have been regarded as highly tissue-specific. However, recent evidence has demonstrated that many enhancers are pleiotropic, with activity in multiple developmental contexts. Yet, the extent and impact of pleiotropy remain largely unexplored. In this study we analyzed active enhancers across human organs based on the analysis of both eRNA transcription (FANTOM5 consortium data sets) and chromatin architecture (ENCODE consortium data sets). We show that pleiotropic enhancers are pervasive in the human genome and that most enhancers active in a particular organ are also active in other organs. In addition, our analysis suggests that the proportion of context-specific enhancers of a given organ is explained, at least in part, by the proportion of context-specific genes in that same organ. The notion that such a high proportion of human enhancers can be pleiotropic suggests that small regions of regulatory DNA contain abundant regulatory information and that these regions evolve under important evolutionary constraints. © The Author(s) 2022. Published by Oxford University Press on behalf of Society for Molecular Biology and Evolution. DOI: 10.1093/gbe/evac071 PMCID: PMC9156028 PMID: 35552697 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/24920384
1. Evol Dev. 2014 Jul-Aug;16(4):207-23. doi: 10.1111/ede.12084. Epub 2014 Jun 11. Functional analysis of limb transcriptional enhancers in the mouse. Nolte MJ(1), Wang Y, Deng JM, Swinton PG, Wei C, Guindani M, Schwartz RJ, Behringer RR. Author information: (1)Graduate Program in Genes and Development, University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA; Department of Genetics, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA. Erratum in Evol Dev. 2014 Nov;16(6):397. Transcriptional enhancers are genomic sequences bound by transcription factors that act together with basal transcriptional machinery to regulate gene transcription. Several high-throughput methods have generated large datasets of tissue-specific enhancer sequences with putative roles in developmental processes. However, few enhancers have been deleted from the genome to determine their roles in development. To understand the roles of two enhancers active in the mouse embryonic limb bud we deleted them from the genome. Although the genes regulated by these enhancers are unknown, they were selected because they were identified in a screen for putative limb bud-specific enhancers associated with p300, an acetyltransferase that participates in protein complexes that promote active transcription, and because the orthologous human enhancers (H1442 and H280) drive distinct lacZ expression patterns in limb buds of embryonic day (E) 11.5 transgenic mice. We show that the orthologous mouse sequences, M1442 and M280, regulate dynamic expression in the developing limb. Although significant transcriptional differences in enhancer-proximal genes in embryonic limb buds accompany the deletion of M1442 and M280 no gross limb malformations during embryonic development were observed, demonstrating that M1442 and M280 are not required for mouse limb development. However, M280 is required for the development and/or maintenance of body size; M280 mice are significantly smaller than controls. M280 also harbors an "ultraconserved" sequence that is identical between human, rat, and mouse. This is the first report of a phenotype resulting from the deletion of an ultraconserved element. These studies highlight the importance of determining enhancer regulatory function by experiments that manipulate them in situ and suggest that some of an enhancer's regulatory capacities may be developmentally tolerated rather than developmentally required. © 2014 Wiley Periodicals, Inc. DOI: 10.1111/ede.12084 PMCID: PMC4130292 PMID: 24920384 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/23651551
1. FEBS Lett. 2013 Jun 27;587(13):1840-7. doi: 10.1016/j.febslet.2013.04.027. Epub 2013 May 4. Communication of genome regulatory elements in a folded chromosome. Razin SV(1), Gavrilov AA, Ioudinkova ES, Iarovaia OV. Author information: (1)Institute of Gene Biology of the Russian Academy of Sciences, 119334 Moscow, Russia. sergey.v.razin@inbox.ru The most popular model of gene activation by remote enhancers postulates that the enhancers interact directly with target promoters via the looping of intervening DNA fragments. This interaction is thought to be necessary for the stabilization of the Pol II pre-initiation complex and/or for the transfer of transcription factors and Pol II, which are initially accumulated at the enhancer, to the promoter. The direct interaction of enhancer(s) and promoter(s) is only possible when these elements are located in close proximity within the nuclear space. Here, we discuss the molecular mechanisms for maintaining the close proximity of the remote regulatory elements of the eukaryotic genome. The models of an active chromatin hub (ACH) and an active nuclear compartment are considered, focusing on the role of chromatin folding in juxtaposing remote DNA sequences. The interconnection between the functionally dependent architecture of the interphase chromosome and nuclear compartmentalization is also discussed. Copyright © 2013 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved. DOI: 10.1016/j.febslet.2013.04.027 PMID: 23651551 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/32952117
1. Yi Chuan. 2020 Sep 20;42(9):817-831. doi: 10.16288/j.yczz.20-097. The identification of enhancers and its application in cancer studies. Liu Q(1), Li CY(1)(2). Author information: (1)Beijing Advanced Innovation Center for Big Data-Based Precision Medicine & School of Medical Sciences and Engineering, Beihang University, Beijing 100191, China. (2)Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology, Beijing 100191, China. Enhancers are a type of cis-acting DNA elements that enhance transcriptional activity of target genes. However, the uncertainty in the orientation and distance between enhancers and target genes could post significant difficulties in identifying the target genes and the regulatory mechanisms of the enhancers. Numerous studies have shown that the mutations and/or abnormalities in the functions of enhancers are associated with development of diseases. A few studies have reported that enhancers could activate cancer development or drug resistance by promoting the expression of target genes. At present, enhancers involved in carcinogenesis and drug resistance have not been fully identified, and the underlying mechanism are still largely unknown. This paper summarizes the main methods used in identifying and characterizing enhancers and analyzing the regulatory mechanism at the genome-wide level. It further reviews the recent research progress of enhancers in cancer diagnosis, treatment, and the underlying mechanism during carcinogenesis, thereby providing a reference for the screening of these enhancers involved in carcinogenesis and drug resistance and exploring their regulatory mechanisms of target genes. It also provides a new perspective for improving the diagnosis of cancer and insights for formulating cancer therapeutic strategies. Publisher: 增强子是一类增强靶基因转录活性的DNA顺式作用元件。但是增强子与靶基因的方向和距离不确定,大大增加了研究增强子调控的靶基因及其作用机制的困难。已有大量研究显示,增强子的突变或功能异常与疾病发生发展相关;仅有少量研究报道增强子通过促进靶基因的表达,引发癌症或产生抗药性。目前与癌症发生发展和在癌症治疗过程中抗药性产生相关的增强子尚未得到充分鉴定,这些增强子的调控机制也未得到充分解析。本文对目前可在全基因组水平上预测和鉴定增强子以及解析增强子调控机制的方法进行总结和对比,并对近几年增强子在肿瘤诊断、治疗和发生发展机制中的研究进展进行综述。期望本文为筛选与癌症发生发展相关的增强子和解析这些增强子的调控机制提供参考,为提高癌症的诊断和制定癌症的治疗策略提供新的视角。. DOI: 10.16288/j.yczz.20-097 PMID: 32952117 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34968246
1. Epigenomes. 2021 Oct 4;5(4):21. doi: 10.3390/epigenomes5040021. Role of Enhancers in Development and Diseases. Maurya SS(1). Author information: (1)Department of Pediatrics, Division of Pediatric Hematology and Oncology, Department of Developmental Biology, School of Medicine, Washington University in St. Louis, 660 South Euclid Avenue, St. Louis, MO 63110, USA. Enhancers are cis-regulatory elements containing short DNA sequences that serve as binding sites for pioneer/regulatory transcription factors, thus orchestrating the regulation of genes critical for lineage determination. The activity of enhancer elements is believed to be determined by transcription factor binding, thus determining the cell state identity during development. Precise spatio-temporal control of the transcriptome during lineage specification requires the coordinated binding of lineage-specific transcription factors to enhancers. Thus, enhancers are the primary determinants of cell identity. Numerous studies have explored the role and mechanism of enhancers during development and disease, and various basic questions related to the functions and mechanisms of enhancers have not yet been fully answered. In this review, we discuss the recently published literature regarding the roles of enhancers, which are critical for various biological processes governing development. Furthermore, we also highlight that altered enhancer landscapes provide an essential context to understand the etiologies and mechanisms behind numerous complex human diseases, providing new avenues for effective enhancer-based therapeutic interventions. DOI: 10.3390/epigenomes5040021 PMCID: PMC8715447 PMID: 34968246 Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/34897508
1. Nucleic Acids Res. 2022 Jan 11;50(1):92-107. doi: 10.1093/nar/gkab1177. Dynamic modulation of enhancer responsiveness by core promoter elements in living Drosophila embryos. Yokoshi M(1), Kawasaki K(1), Cambón M(2), Fukaya T(1)(3). Author information: (1)Laboratory of Transcription Dynamics, Research Center for Biological Visualization, Institute for Quantitative Biosciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan. (2)Applied Mathematics Department, University of Granada, Granada, Spain. (3)Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan. Regulatory interactions between enhancers and core promoters are fundamental for the temporal and spatial specificity of gene expression in development. The central role of core promoters is to initiate productive transcription in response to enhancer's activation cues. However, it has not been systematically assessed how individual core promoter elements affect the induction of transcriptional bursting by enhancers. Here, we provide evidence that each core promoter element differentially modulates functional parameters of transcriptional bursting in developing Drosophila embryos. Quantitative live imaging analysis revealed that the timing and the continuity of burst induction are common regulatory steps on which core promoter elements impact. We further show that the upstream TATA also affects the burst amplitude. On the other hand, Inr, MTE and DPE mainly contribute to the regulation of the burst frequency. Genome editing analysis of the pair-rule gene fushi tarazu revealed that the endogenous TATA and DPE are both essential for its correct expression and function during the establishment of body segments in early embryos. We suggest that core promoter elements serve as a key regulatory module in converting enhancer activity into transcription dynamics during animal development. © The Author(s) 2021. Published by Oxford University Press on behalf of Nucleic Acids Research. DOI: 10.1093/nar/gkab1177 PMCID: PMC8754644 PMID: 34897508 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/24599251
1. Nat Struct Mol Biol. 2014 Mar;21(3):210-9. doi: 10.1038/nsmb.2784. Enhancer biology and enhanceropathies. Smith E(1), Shilatifard A(1). Author information: (1)Stowers Institute for Medical Research, Kansas City, Missouri, USA. Enhancers are cis-regulatory elements that enable precise spatiotemporal patterns of gene expression during development and are notable for being able to function at large distances from their target genes. Such regulatory elements often bypass intervening genes and typically comprise binding sites for multiple transcription factors that can also be transcribed by RNA polymerase II (Pol II) to produce noncoding enhancer RNAs (eRNAs). Genome-wide analyses have revealed chromatin signatures of enhancers, such as the enrichment for monomethylation of histone H3 lysine 4 (H3K4me1) and the acetylation or methylation of histone H3 lysine 27 (H3K27). Enhancer signatures have been used to describe the transitions of these regulatory elements from inactive to primed and from activated to decommissioned states during development. New mutations of enhancer sequences and of the protein factors regulating enhancer function in human disease continue to be identified, contributing to a growing class of 'enhanceropathies'. DOI: 10.1038/nsmb.2784 PMID: 24599251 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/32635644
1. Cells. 2020 Jul 5;9(7):1620. doi: 10.3390/cells9071620. Molecular Basis of the Function of Transcriptional Enhancers. Ibragimov AN(1)(2), Bylino OV(1), Shidlovskii YV(1)(3). Author information: (1)Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia. (2)Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia. (3)I.M. Sechenov First Moscow State Medical University, 8, bldg. 2 Trubetskaya St., 119048 Moscow, Russia. Transcriptional enhancers are major genomic elements that control gene activity in eukaryotes. Recent studies provided deeper insight into the temporal and spatial organization of transcription in the nucleus, the role of non-coding RNAs in the process, and the epigenetic control of gene expression. Thus, multiple molecular details of enhancer functioning were revealed. Here, we describe the recent data and models of molecular organization of enhancer-driven transcription. DOI: 10.3390/cells9071620 PMCID: PMC7407508 PMID: 32635644 [Indexed for MEDLINE] Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/27863239
1. Cell. 2016 Nov 17;167(5):1170-1187. doi: 10.1016/j.cell.2016.09.018. Ever-Changing Landscapes: Transcriptional Enhancers in Development and Evolution. Long HK(1), Prescott SL(2), Wysocka J(3). Author information: (1)Department of Chemical and Systems Biology, Stanford School of Medicine, Stanford University, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford University, Stanford, CA 94305, USA. (2)Department of Chemical and Systems Biology, Stanford School of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Developmental Biology, Stanford School of Medicine, Stanford University, Stanford, CA 94305, USA. (3)Department of Chemical and Systems Biology, Stanford School of Medicine, Stanford University, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Developmental Biology, Stanford School of Medicine, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford School of Medicine, Stanford University, Stanford, CA 94305, USA. Electronic address: wysocka@stanford.edu. A class of cis-regulatory elements, called enhancers, play a central role in orchestrating spatiotemporally precise gene-expression programs during development. Consequently, divergence in enhancer sequence and activity is thought to be an important mediator of inter- and intra-species phenotypic variation. Here, we give an overview of emerging principles of enhancer function, current models of enhancer architecture, genomic substrates from which enhancers emerge during evolution, and the influence of three-dimensional genome organization on long-range gene regulation. We discuss intricate relationships between distinct elements within complex regulatory landscapes and consider their potential impact on specificity and robustness of transcriptional regulation. Copyright © 2016 Elsevier Inc. All rights reserved. DOI: 10.1016/j.cell.2016.09.018 PMCID: PMC5123704 PMID: 27863239 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/25719310
1. Biol Chem. 2015 Apr;396(4):311-27. doi: 10.1515/hsz-2014-0303. Enhancers, enhancers - from their discovery to today's universe of transcription enhancers. Schaffner W. Transcriptional enhancers are short (200-1500 base pairs) DNA segments that are able to dramatically boost transcription from the promoter of a target gene. Originally discovered in simian virus 40 (SV40), a small DNA virus, transcription enhancers were soon also found in immunoglobulin genes and other cellular genes as key determinants of cell-type-specific gene expression. Enhancers can exert their effect over long distances of thousands, even hundreds of thousands of base pairs, either from upstream, downstream, or from within a transcription unit. The number of enhancers in eukaryotic genomes correlates with the complexity of the organism; a typical mammalian gene is likely controlled by several enhancers to fine-tune its expression at different developmental stages, in different cell types and in response to different signaling cues. Here, I provide a personal account of how enhancers were discovered more than 30 years ago, and also address the amazing development of the field since then. DOI: 10.1515/hsz-2014-0303 PMID: 25719310 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/30780195
1. Dev Growth Differ. 2019 Jun;61(5):343-352. doi: 10.1111/dgd.12597. Epub 2019 Feb 19. Dynamics of transcriptional enhancers and chromosome topology in gene regulation. Yokoshi M(1), Fukaya T(1)(2). Author information: (1)Institute for Quantitative Biosciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan. (2)Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan. Transcriptional enhancers are regulatory DNAs that instruct when and where genes should be transcribed in response to a variety of intrinsic and external signals. They contain a cluster of binding sites for sequence-specific transcription factors and co-activators to determine the spatiotemporal specificity of gene activities during development. Enhancers are often positioned in distal locations from their target promoters. In some cases, they work over a million base pairs or more. In the traditional view, enhancers have been thought to stably interact with promoters in a targeted manner. However, quantitative imaging studies provide a far more dynamic picture of enhancer action. Moreover, recent Hi-C methods suggest that regulatory interactions are dynamically regulated by the higher-order chromosome topology. In this review, we summarize the emerging findings in the field and propose that assembly of "transcription hubs" in the context of 3D genome structure plays an important role in transcriptional regulation. © 2019 The Authors. Development, Growth & Differentiation published by John Wiley & Sons Australia, Ltd on behalf of Japanese Society of Developmental Biologists. DOI: 10.1111/dgd.12597 PMCID: PMC6850047 PMID: 30780195 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35868358
1. J Control Release. 2022 Sep;349:1045-1051. doi: 10.1016/j.jconrel.2022.05.061. Epub 2022 Sep 8. Suprachoroidal delivery enables targeting, localization and durability of small molecule suspensions. Kansara VS(1), Hancock SE(1), Muya LW(1), Ciulla TA(2). Author information: (1)Clearside Biomedical Inc., 900 North Point Parkway, Suite 200, Alpharetta, GA 30005, United States of America. (2)Clearside Biomedical Inc., 900 North Point Parkway, Suite 200, Alpharetta, GA 30005, United States of America. Electronic address: thomas.ciulla@clearsidebio.com. Drug delivery to the suprachoroidal space (SCS®) has become a clinical reality after the 2021 FDA approval of CLS-TA, a triamcinolone acetonide injectable suspension for suprachoroidal use (XIPERE®), administered via a microneedle-based device, the SCS Microinjector®. Suprachoroidal (SC) delivery facilitates targeting, compartmentalization, and durability of small molecule suspensions, thereby potentially addressing some of the efficacy, safety, and treatment burden limitations of current retinal therapies. Herein, the design features of the SCS Microinjector are reviewed, along with the biomechanics of SC drug delivery. Also presented are preclinical evaluations of SC small molecule suspensions from 4 different therapeutic classes (plasma kallikrein inhibitor, receptor tyrosine kinase inhibitor, corticosteroid, complement factor D inhibitor), highlighting their potential for durability, targeted compartmentalization, and acceptable safety profiles following microinjector-based SC delivery. The clinical evaluations of the safety, tolerability and efficacy of SC delivered triamcinolone further supports potential of SC small molecule suspensions as a clinically viable strategy for the treatment of chorioretinal diseases. Also highlighted are current limitations, key pharmacological considerations, and future opportunities to optimize the SC microinjector platform for safe, effective, and potentially long-acting drug delivery for the treatment of chorioretinal disorders. Copyright © 2022 The Author(s). Published by Elsevier B.V. All rights reserved. DOI: 10.1016/j.jconrel.2022.05.061 PMID: 35868358 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35562158
1. Indian J Pathol Microbiol. 2022 May;65(Supplement):S259-S270. doi: 10.4103/ijpm.ijpm_1074_21. Genetics and muscle pathology in the diagnosis of muscular dystrophies: An update. Narasimhaiah D(1), Uppin MS(2), Ranganath P(3). Author information: (1)Department of Pathology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India. (2)Department of Pathology, Nizam's Institute of Medical Sciences, Punjagutta, Hyderabad, Telangana, India. (3)Department of Medical Genetics, Nizam's Institute of Medical Sciences, Punjagutta, Hyderabad, Telangana, India. Muscular dystrophies are a clinically and genetically heterogeneous group of disorders involving the skeletal muscles. They have a progressive clinical course and are characterized by muscle fiber degeneration. Congenital muscular dystrophies (CMD) include dystroglycanopathies, merosin-deficient CMD, collagen VI-deficient CMD, SELENON-related rigid spine muscular dystrophy, and LMNA-related CMD. Childhood and adult-onset muscular dystrophies include dystrophinopathies, limb-girdle muscular dystrophies, Emery-Dreifuss muscular dystrophy, facioscapulohumeral muscular dystrophy, and myotonic dystrophy. Traditionally, muscle biopsy and histopathology along with special pathology techniques such as immunohistochemistry or immunoblotting were used for the diagnosis of muscular dystrophies. However, recent advances in molecular genetic testing, especially the next-generation sequencing technology, have revolutionized the diagnosis of muscular dystrophies. Identification of the underlying genetic basis helps in appropriate management and prognostication of the affected individual and genetic counseling of the family. In addition, identification of the exact disease-causing mutations is necessary for accurate prenatal genetic testing and carrier testing, to prevent recurrence in the family. Mutation identification is also essential for initiating mutation-specific therapies (which have been developed recently, especially for Duchenne muscular dystrophy) and for enrolment of patients into ongoing therapeutic clinical trials. The 'genetic testing first' approach has now become the norm in most centers. Nonetheless, muscle biopsy-based testing still has an important role to play, especially for cases where genetic testing is negative or inconclusive for the etiology. DOI: 10.4103/ijpm.ijpm_1074_21 PMID: 35562158 [Indexed for MEDLINE] Conflict of interest statement: None
http://www.ncbi.nlm.nih.gov/pubmed/27340611
1. Expert Opin Orphan Drugs. 2016;4(2):169-183. doi: 10.1517/21678707.2016.1124039. Epub 2015 Dec 17. Prospect of gene therapy for cardiomyopathy in hereditary muscular dystrophy. Yue Y(1), Binalsheikh IM(2), Leach SB(3), Domeier TL(4), Duan D(5). Author information: (1)Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri. (2)Department of Child Health, School of Medicine, University of Missouri. (3)Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri. (4)Department of Medical Physiology and Pharmacology, School of Medicine, University of Missouri. (5)Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri; Department of Neurology, School of Medicine, University of Missouri. INTRODUCTION: Cardiac involvement is a common feature in muscular dystrophies. It presents as heart failure and/or arrhythmia. Traditionally, dystrophic cardiomyopathy is treated with symptom-relieving medications. Identification of disease-causing genes and investigation on pathogenic mechanisms have opened new opportunities to treat dystrophic cardiomyopathy with gene therapy. Replacing/repairing the mutated gene and/or targeting the pathogenic process/mechanisms using alternative genes may attenuate heart disease in muscular dystrophies. AREAS COVERED: Duchenne muscular dystrophy is the most common muscular dystrophy. Duchenne cardiomyopathy has been the primary focus of ongoing dystrophic cardiomyopathy gene therapy studies. Here, we use Duchenne cardiomyopathy gene therapy to showcase recent developments and to outline the path forward. We also discuss gene therapy status for cardiomyopathy associated with limb-girdle and congenital muscular dystrophies, and myotonic dystrophy. EXPERT OPINION: Gene therapy for dystrophic cardiomyopathy has taken a slow but steady path forward. Preclinical studies over the last decades have addressed many fundamental questions. Adeno-associated virus-mediated gene therapy has significantly improved the outcomes in rodent models of Duchenne and limb girdle muscular dystrophies. Validation of these encouraging results in large animal models will pave the way to future human trials. DOI: 10.1517/21678707.2016.1124039 PMCID: PMC4914135 PMID: 27340611 Conflict of interest statement: Financial and competing interests disclosure The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript.
http://www.ncbi.nlm.nih.gov/pubmed/19555515
1. Expert Rev Mol Med. 2009 Jun 25;11:e18. doi: 10.1017/S1462399409001100. Emerging strategies for cell and gene therapy of the muscular dystrophies. Muir LA(1), Chamberlain JS. Author information: (1)Program in Molecular and Cellular Biology, University of Washington, Seattle, Washington 98195, USA. The muscular dystrophies are a heterogeneous group of over 40 disorders that are characterised by muscle weakness and wasting. The most common are Duchenne muscular dystrophy and Becker muscular dystrophy, which result from mutations within the gene encoding dystrophin; myotonic dystrophy type 1, which results from an expanded trinucleotide repeat in the myotonic dystrophy protein kinase gene; and facioscapulohumeral dystrophy, which is associated with contractions in the subtelomeric region of human chromosome 1. Currently the only treatments involve clinical management of symptoms, although several promising experimental strategies are emerging. These include gene therapy using adeno-associated viral, lentiviral and adenoviral vectors and nonviral vectors, such as plasmid DNA. Exon-skipping and cell-based therapies have also shown promise in the effective treatment and regeneration of dystrophic muscle. The availability of numerous animal models for Duchenne muscular dystrophy has enabled extensive testing of a wide range of therapeutic approaches for this type of disorder. Consequently, we focus here on the therapeutic developments for Duchenne muscular dystrophy as a model of the types of approaches being considered for various types of dystrophy. We discuss the advantages and limitations of each therapeutic strategy, as well as prospects and recent successes in the context of future clinical applications. DOI: 10.1017/S1462399409001100 PMCID: PMC4890545 PMID: 19555515 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/24213305
1. J Trop Pediatr. 2014 Apr;60(2):112-7. doi: 10.1093/tropej/fmt090. Epub 2013 Nov 7. Genetic diagnosis of Duchenne and Becker muscular dystrophy using multiplex ligation-dependent probe amplification in Rwandan patients. Uwineza A(1), Hitayezu J, Murorunkwere S, Ndinkabandi J, Kalala Malu CK, Caberg JH, Dideberg V, Bours V, Mutesa L. Author information: (1)Center for Medical Genetics, Department of Molecular Biology & Cytogenetics, Faculty of Medicine, National University of Rwanda, PO Box 30-Butare, Rwanda. Duchenne and Becker muscular dystrophies are the most common clinical forms of muscular dystrophies. They are genetically X-linked diseases caused by a mutation in the dystrophin (DMD) gene. A genetic diagnosis was carried out in six Rwandan patients presenting a phenotype of Duchenne and Becker muscular dystrophies and six asymptomatic female carrier relatives using multiplex ligation-dependent probe amplification (MLPA). Our results revealed deletion of the exons 48-51 in one patient, an inherited deletion of the exons 8-21 in two brothers and a de novo deletion of the exons 46-50 in the fourth patient. No copy number variation was found in two patients. Only one female carrier presented exon deletion in the DMD gene. This is the first cohort of genetic analysis in Rwandan patients affected by Duchenne and Becker muscular dystrophies. This report confirmed that MLPA assay can be easily implemented in low-income countries. DOI: 10.1093/tropej/fmt090 PMID: 24213305 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/3728255
1. Am Fam Physician. 1986 Jul;34(1):123-8. The muscular dystrophies. Seiler J, Bope ET. Duchenne muscular dystrophy, the most common childhood-onset muscular dystrophy, is X-linked and is associated with cardiac and mental abnormalities. Becker's muscular dystrophy is similar to but milder than Duchenne muscular dystrophy. The rare facioscapulohumeral muscular dystrophy has an autosomal dominant mode of transmission. Myotonic muscular dystrophy is the most common of the adult-onset muscular dystrophies. Treatable diseases that must be excluded include polymyositis, potassium disorders and endocrine abnormalities. PMID: 3728255 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/24305447
1. Continuum (Minneap Minn). 2013 Dec;19(6 Muscle Disease):1535-70. doi: 10.1212/01.CON.0000440659.41675.8b. The muscular dystrophies. Wicklund MP. Erratum in Continuum (Minneap Minn). 2014 Jun;20(3 Neurology of Systemic Disease):520. PURPOSE OF REVIEW: With transition to the genetic era, the number of muscular dystrophies has grown significantly, but so too has our understanding of their pathogenic underpinnings. Clinical features associated with each muscular dystrophy still guide us to the diagnosis. However, improved diagnostic abilities refine and expand phenotypic and genotypic correlates. This article discusses the epidemiology, clinical features, and diagnosis of these disorders. RECENT FINDINGS: Some important recent advancements include (1) a much greater understanding of the pathogenetic pathways underlying facioscapulohumeral muscular dystrophy and myotonic dystrophy type 1; (2) the publication of diagnostic and treatment guidelines for Duchenne muscular dystrophy; and (3) further clarification of the many genetic muscle disorders presenting a limb-girdle pattern of weakness. SUMMARY: Muscular dystrophies are genetic, progressive, degenerative disorders with the primary symptom of muscle weakness. Duchenne, Becker, facioscapulohumeral, and myotonic muscular dystrophies are most prevalent and tend to have distinctive features helpful in diagnosis. The limb-girdle, Emery-Dreifuss, and oculopharyngeal muscular dystrophies are less common but often may also be diagnosed on the basis of phenotype. Researchers hope to help patients with future discoveries effective in slowing or halting disease progression, reversing or preventing underlying mechanisms, and repairing previously damaged muscle. DOI: 10.1212/01.CON.0000440659.41675.8b PMCID: PMC10564029 PMID: 24305447 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/31794464
1. Continuum (Minneap Minn). 2019 Dec;25(6):1640-1661. doi: 10.1212/CON.0000000000000792. Congenital Muscular Dystrophy and Congenital Myopathy. Butterfield RJ. PURPOSE OF REVIEW: Congenital muscular dystrophies and congenital myopathies are a heterogeneous group of disorders resulting in hypotonia, muscle weakness, and dystrophic or myopathic features on muscle biopsy. This article summarizes the clinical and genetic aspects of these disorders. RECENT FINDINGS: Historically, diagnoses of congenital muscular dystrophy and congenital myopathy have been made by clinical features and histopathology; however, recent advances in genetics have changed diagnostic practice by relying more heavily on genetic findings. This article reviews the clinical and genetic features of the most common congenital muscular dystrophies including laminin subunit alpha 2 (LAMA2)-related (merosin deficient), collagen VI-related, and α-dystroglycan-related congenital muscular dystrophies and reviews the most common congenital myopathies including nemaline rod, core, and centronuclear myopathies. With the increasing accessibility of genetic testing, the number of genes found to be associated with these disorders has increased dramatically. A wide spectrum of severity and onset (from birth to adulthood) exist across all subtypes. Progression and other features are variable depending on the subtype and severity of the specific genetic mutation. SUMMARY: Congenital muscular dystrophy and congenital myopathy are increasingly recognized disorders. A growing appreciation for the breadth of phenotypic variability and overlap between established subtypes has challenged long-standing phenotypic and histopathologic classifications of these disorders but has driven a greater understanding of pathogenesis and opened the door to the development of novel treatments. DOI: 10.1212/CON.0000000000000792 PMID: 31794464 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/30215857
1. Compr Physiol. 2018 Sep 14;8(4):1313-1356. doi: 10.1002/cphy.c170052. Immunobiology of Inherited Muscular Dystrophies. Tidball JG(1), Welc SS(2), Wehling-Henricks M(3). Author information: (1)Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, California, USA. (2)Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA. (3)Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA. The immune response to acute muscle damage is important for normal repair. However, in chronic diseases such as many muscular dystrophies, the immune response can amplify pathology and play a major role in determining disease severity. Muscular dystrophies are inheritable diseases that vary tremendously in severity, but share the progressive loss of muscle mass and function that can be debilitating and lethal. Mutations in diverse genes cause muscular dystrophy, including genes that encode proteins that maintain membrane strength, participate in membrane repair, or are components of the extracellular matrix or the nuclear envelope. In this article, we explore the hypothesis that an important feature of many muscular dystrophies is an immune response adapted to acute, infrequent muscle damage that is misapplied in the context of chronic injury. We discuss the involvement of the immune system in the most common muscular dystrophy, Duchenne muscular dystrophy, and show that the immune system influences muscle death and fibrosis as disease progresses. We then present information on immune cell function in other muscular dystrophies and show that for many muscular dystrophies, release of cytosolic proteins into the extracellular space may provide an initial signal, leading to an immune response that is typically dominated by macrophages, neutrophils, helper T-lymphocytes, and cytotoxic T-lymphocytes. Although those features are similar in many muscular dystrophies, each muscular dystrophy shows distinguishing features in the magnitude and type of inflammatory response. These differences indicate that there are disease-specific immunomodulatory molecules that determine response to muscle cell damage caused by diverse genetic mutations. © 2018 American Physiological Society. Compr Physiol 8:1313-1356, 2018. Copyright © 2018 American Physiological Society. All rights reserved. DOI: 10.1002/cphy.c170052 PMCID: PMC7769418 PMID: 30215857 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/27734165
1. J Neurol. 2016 Dec;263(12):2528-2537. doi: 10.1007/s00415-016-8300-3. Epub 2016 Oct 12. Medication adherence in patients with myotonic dystrophy and facioscapulohumeral muscular dystrophy. Fitzgerald BP(1), Conn KM(2), Smith J(2), Walker A(2), Parkhill AL(2), Hilbert JE(3), Luebbe EA(3), Moxley RT III(3). Author information: (1)Wegmans School of Pharmacy, St. John Fisher College, 3690 East Avenue, Rochester, NY, 14618, USA. bpf08248@sjfc.edu. (2)Wegmans School of Pharmacy, St. John Fisher College, 3690 East Avenue, Rochester, NY, 14618, USA. (3)Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA. Myotonic dystrophy (DM) and facioscapulohumeral muscular dystrophy (FSHD) are the two most common adult muscular dystrophies and have progressive and often disabling manifestations. Higher levels of medication adherence lead to better health outcomes, especially important to patients with DM and FSHD because of their multisystem manifestations and complexity of care. However, medication adherence has not previously been studied in a large cohort of DM type 1 (DM1), DM type 2 (DM2), and FSHD patients. The purpose of our study was to survey medication adherence and disease manifestations in patients enrolled in the NIH-supported National DM and FSHD Registry. The study was completed by 110 DM1, 49 DM2, and 193 FSHD patients. Notable comorbidities were hypertension in FSHD (44 %) and DM2 (37 %), gastroesophageal reflux disease in DM1 (24 %) and DM2 (31 %) and arrhythmias (29 %) and thyroid disease (20 %) in DM1. Each group reported high levels of adherence based on regimen complexity, medication costs, health literacy, side effect profile, and their beliefs about treatment. Only dysphagia in DM1 was reported to significantly impact medication adherence. Approximately 35 % of study patients reported polypharmacy (taking 6 or more medications). Of the patients with polypharmacy, the DM1 cohort was significantly younger (mean 55.0 years) compared to DM2 (59.0 years) and FSHD (63.2 years), and had shorter disease duration (mean 26 years) compared to FSHD (26.8 years) and DM2 (34.8 years). Future research is needed to assess techniques to ease pill swallowing in DM1 and to monitor polypharmacy and potential drug interactions in DM and FSHD. DOI: 10.1007/s00415-016-8300-3 PMCID: PMC5112113 PMID: 27734165 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/23733645
1. Compr Physiol. 2011 Jul;1(3):1353-63. doi: 10.1002/cphy.c100062. Exercise and muscular dystrophy: implications and analysis of effects on musculoskeletal and cardiovascular systems. Barnabei MS(1), Martindale JM, Townsend D, Metzger JM. Author information: (1)Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA. The muscular dystrophies are a heterogeneous collection of progressive, inherited diseases of muscle weakness and degeneration. Although these diseases can vary widely in their etiology and presentation, nearly all muscular dystrophies cause exercise intolerance to some degree. Here, we focus on Duchenne muscular dystrophy (DMD), the most common form of muscular dystrophy, as a paradigm for the effects of muscle disease on exercise capacity. First described in the mid-1800s, DMD is a rapidly progressive and lethal muscular dystrophy caused by mutations in the dystrophin gene. Dystrophin is a membrane-associated cytoskeletal protein, the loss of which causes numerous cellular defects including mechanical instability of the sarcolemma, increased influx of extracellular calcium, and cell signaling defects. Here, we discuss the physiological basis for exercise intolerance in DMD, focusing on the molecular and cellular defects caused by loss of dystrophin and how these manifest as organ-level dysfunction and reduced exercise capacity. The main focus of this article is the defects present in dystrophin-deficient striated muscle. However, discussion regarding the effects of dystrophin loss on other tissues, including vascular smooth muscle is also included. Collectively, the goal of this article is to summarize the current state of knowledge regarding the mechanistic basis for exercise intolerance in DMD, which may serve as an archetype for other muscular dystrophies and diseases of muscle wasting. © 2011 American Physiological Society. DOI: 10.1002/cphy.c100062 PMID: 23733645 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/32095889
1. Lung. 2020 Apr;198(2):371-375. doi: 10.1007/s00408-020-00340-7. Epub 2020 Feb 24. Peak Cough Flow in Children with Neuromuscular Disorders. Kotwal N(1)(2), Shukla PJ(3), Perez GF(4)(5). Author information: (1)Division of Pulmonology and Allergy, Department of Pediatrics, University of Maryland, Maryland, 737 W Lombard St, Ste 314, Baltimore, MD, 21201, USA. nkotwal@som.umaryland.edu. (2)Division of Pulmonary and Sleep Medicine, Children's National Hospital, 111 Michigan Ave, NW, Washington, DC, 20010, USA. nkotwal@som.umaryland.edu. (3)Department of Pediatrics, MedStar Georgetown University Hospital, 3800 Reservoir Rd, NW, Washington, DC, 20007, USA. (4)Division of Pulmonary and Sleep Medicine, Children's National Hospital, 111 Michigan Ave, NW, Washington, DC, 20010, USA. (5)Department of Pediatrics, Oishei Children's Hospital, Jacobs School of Medicine and Biomedical Sciences, University of Buffalo, New York, NY, 14203, USA. PURPOSE: Patients with neuromuscular disease (NMD) experience weakened cough due to progressive respiratory muscle weakness. Peak cough flow (PCF) measurements derived from adult populations are used to recommend initiation of assisted cough therapies. The objective of this study was to characterize PCF values among pediatric patients with NMD. METHODS: Retrospective chart review was performed for patients seen in the multidisciplinary pediatric muscular dystrophy clinic from 2010 to 2016. Clinical and demographic variables included age, gender, ambulation status, and PCF measurements. RESULTS: 366 patients with an established diagnosis of NMD (median age 11.8 years) were included in this study. 102 (27.8%) out of the 366 patients were affected by Duchenne muscular dystrophy (DMD), 42 (11.5%) by congenital muscular dystrophy (CMD), 42 (11.5%) by Charcot Marie Tooth disease (CMT) and 24 (6.5%) by Becker's muscular dystrophy (BMD). The mean PCF values in DMD (255.8 L/min) and CMD (249.1 L/min) were lower than CMT (321.5 L/min) with p-values of 0.007 and 0.02, respectively. The mean PCF of BMD (333.3 L/min) was higher than that of DMD and CMD but the difference was not statistically significant. PCFs were not statistically different between ambulatory and non-ambulatory status (263.0 L/min versus 290.8 L/min, p = 0.12). Children under 10 years of age had lower PCF relative to older subjects (179.5 L/min versus 300.9 L/min, p < 0.0001). CONCLUSION: Baseline PCF values in young children are below the adult-specific values suggested for starting assisted cough techniques. Further longitudinal trials are required to derive pediatric-specific reference values for PCF in patients with NMD. DOI: 10.1007/s00408-020-00340-7 PMID: 32095889 [Indexed for MEDLINE]