Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
J Med Chem ; 65(18): 12417-12426, 2022 09 22.
Article in English | MEDLINE | ID: mdl-36099320

ABSTRACT

Trinucleotide repeat diseases such as myotonic dystrophy type 1 (DM1) and Huntington's disease (HD) are caused by expanded DNA repeats that can be used as templates to synthesize their own inhibitors. Because it would be particularly advantageous to reversibly assemble multivalent nucleic acid-targeting agents in situ, we sought to develop a target-guided screen that uses dynamic covalent chemistry to identify multitarget inhibitors. We report the synthesis of a library of amine- or aldehyde-containing fragments. The assembly of these fragments led to a diverse set of hit combinations that was confirmed by matrix-assisted laser desorption/ionization-mass spectrometry (MALDI-MS) in the presence of DM1 and HD repeat sequences. Of interest for both diseases, the resulting hit combinations inhibited transcription selectively and in a cooperative manner in vitro, with inhibitory concentration (IC50) values in the micromolar range. This dynamic covalent library and screening approach could be applied to identify compounds that reversibly assemble on other nucleic acid targets.


Subject(s)
Aldehydes , Amines , Nucleic Acids , Aldehydes/chemical synthesis , Aldehydes/pharmacology , Amines/chemical synthesis , Amines/pharmacology , Drug Evaluation, Preclinical , Humans , Huntington Disease/genetics , Myotonic Dystrophy/genetics , Nucleic Acids/antagonists & inhibitors , Nucleic Acids/chemistry , Repetitive Sequences, Nucleic Acid , Transcription, Genetic/drug effects
2.
Elife ; 102021 09 14.
Article in English | MEDLINE | ID: mdl-34517941

ABSTRACT

Microsatellite expansions of CCTG repeats in the cellular nucleic acid-binding protein (CNBP) gene leads to accumulation of toxic RNA and have been associated with myotonic dystrophy type 2 (DM2). However, it is still unclear whether the dystrophic phenotype is also linked to CNBP decrease, a conserved CCHC-type zinc finger RNA-binding protein that regulates translation and is required for mammalian development. Here, we show that depletion of Drosophila CNBP in muscles causes ageing-dependent locomotor defects that are correlated with impaired polyamine metabolism. We demonstrate that the levels of ornithine decarboxylase (ODC) and polyamines are significantly reduced upon dCNBP depletion. Of note, we show a reduction of the CNBP-polyamine axis in muscles from DM2 patients. Mechanistically, we provide evidence that dCNBP controls polyamine metabolism through binding dOdc mRNA and regulating its translation. Remarkably, the locomotor defect of dCNBP-deficient flies is rescued by either polyamine supplementation or dOdc1 overexpression. We suggest that this dCNBP function is evolutionarily conserved in vertebrates with relevant implications for CNBP-related pathophysiological conditions.


Subject(s)
Drosophila Proteins/metabolism , Motor Activity/genetics , Motor Activity/physiology , Polyamines/metabolism , RNA-Binding Proteins/metabolism , Animals , Animals, Genetically Modified , Cell Line , Down-Regulation/physiology , Drosophila Proteins/genetics , Drosophila melanogaster , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Muscle, Skeletal/metabolism , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Protein Biosynthesis , Putrescine/pharmacology , RNA Interference , RNA-Binding Proteins/genetics , Spermidine/pharmacology
3.
Biol Rev Camb Philos Soc ; 96(2): 716-730, 2021 04.
Article in English | MEDLINE | ID: mdl-33269537

ABSTRACT

Myotonic dystrophy type 1 (DM1) is the most prevalent form of muscular dystrophy in adults and yet there are currently no treatment options. Although this disease causes multisystemic symptoms, it is mainly characterised by myopathy or diseased muscles, which includes muscle weakness, atrophy, and myotonia, severely affecting the lives of patients worldwide. On a molecular level, DM1 is caused by an expansion of CTG repeats in the 3' untranslated region (3'UTR) of the DM1 Protein Kinase (DMPK) gene which become pathogenic when transcribed into RNA forming ribonuclear foci comprised of auto complementary CUG hairpin structures that can bind proteins. This leads to the sequestration of the muscleblind-like (MBNL) family of proteins, depleting them, and the abnormal stabilisation of CUGBP Elav-like family member 1 (CELF1), enhancing it. Traditionally, DM1 research has focused on this RNA toxicity and how it alters MBNL and CELF1 functions as key splicing regulators. However, other proteins are affected by the toxic DMPK RNA and there is strong evidence that supports various signalling cascades playing an important role in DM1 pathogenesis. Specifically, the impairment of protein kinase B (AKT) signalling in DM1 increases autophagy, apoptosis, and ubiquitin-proteasome activity, which may also be affected in DM1 by AMP-activated protein kinase (AMPK) downregulation. AKT also regulates CELF1 directly, by affecting its subcellular localisation, and indirectly as it inhibits glycogen synthase kinase 3 beta (GSK3ß), which stabilises the repressive form of CELF1 in DM1. Another kinase that contributes to CELF1 mis-regulation, in this case by hyperphosphorylation, is protein kinase C (PKC). Additionally, it has been demonstrated that fibroblast growth factor-inducible 14 (Fn14) is induced in DM1 and is associated with downstream signalling through the nuclear factor κB (NFκB) pathways, associating inflammation with this disease. Furthermore, MBNL1 and CELF1 play a role in cytoplasmic processes involved in DM1 myopathy, altering proteostasis and sarcomere structure. Finally, there are many other elements that could contribute to the muscular phenotype in DM1 such as alterations to satellite cells, non-coding RNA metabolism, calcium dysregulation, and repeat-associated non-ATG (RAN) translation. This review aims to organise the currently dispersed knowledge on the different pathways affected in DM1 and discusses the unexplored connections that could potentially help in providing new therapeutic targets in DM1 research.


Subject(s)
Myotonic Dystrophy , Alternative Splicing , Humans , Muscles/metabolism , Myotonic Dystrophy/genetics , RNA, Messenger/metabolism
4.
Acta Myol ; 39(1): 32-35, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32607478

ABSTRACT

A significant number of sudden death (SD) is observed in myotonic dystrophy (DM1) despite pacemaker implantation and some consider the ICD to be the preferential device in patients with conduction disease. According to the latest guidelines, prophylactic ICD implantation in patients with neuromuscular disorder should follow the same recommendations of non-ischemic dilated cardiomyopathy, being reasonable when pacing is needed. We here report a case of DM1 patient who underwent ICD implantation even in the absence of conduction disturbances on ECG and ventricular dysfunction/fibrosis at cardiac magnetic resonance. The occurrence of syncope, non-sustained ventricular tachycardias at 24-Holter ECG monitoring and a family history of SD resulted associated with ventricular fibrillation inducibility at electrophysiological study, favouring ICD implantation. On our advice, DM1 patient with this association of SD risk factors should be targeted for ICD implantation.


Subject(s)
Bisoprolol/administration & dosage , Death, Sudden, Cardiac/prevention & control , Defibrillators, Implantable , Myotonic Dystrophy , Syncope/diagnosis , Tachycardia, Ventricular , Adrenergic beta-1 Receptor Antagonists/administration & dosage , Adult , Clinical Decision-Making , Electrocardiography, Ambulatory/methods , Electromyography/methods , Electrophysiologic Techniques, Cardiac/methods , Female , Humans , Muscle Weakness/etiology , Myotonic Dystrophy/diagnosis , Myotonic Dystrophy/genetics , Myotonic Dystrophy/physiopathology , Patient Selection , Risk Assessment , Tachycardia, Ventricular/diagnosis , Tachycardia, Ventricular/etiology
6.
Proc Natl Acad Sci U S A ; 116(42): 20991-21000, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31570586

ABSTRACT

A CTG repeat expansion in the DMPK gene is the causative mutation of myotonic dystrophy type 1 (DM1). Transcription of the expanded CTG repeat produces toxic gain-of-function CUG RNA, leading to disease symptoms. A screening platform that targets production or stability of the toxic CUG RNA in a selective manner has the potential to provide new biological and therapeutic insights. A DM1 HeLa cell model was generated that stably expresses a toxic r(CUG)480 and an analogous r(CUG)0 control from DMPK and was used to measure the ratio-metric level of r(CUG)480 versus r(CUG)0. This DM1 HeLa model recapitulates pathogenic hallmarks of DM1, including CUG ribonuclear foci and missplicing of pre-mRNA targets of the muscleblind (MBNL) alternative splicing factors. Repeat-selective screening using this cell line led to the unexpected identification of multiple microtubule inhibitors as hits that selectively reduce r(CUG)480 levels and partially rescue MBNL-dependent missplicing. These results were validated by using the Food and Drug Administration-approved clinical microtubule inhibitor colchicine in DM1 mouse and primary patient cell models. The mechanism of action was found to involve selective reduced transcription of the CTG expansion that we hypothesize to involve the LINC (linker of nucleoskeleton and cytoskeleton) complex. The unanticipated identification of microtubule inhibitors as selective modulators of toxic CUG RNA opens research directions for this form of muscular dystrophy and may shed light on the biology of CTG repeat expansion and inform therapeutic avenues. This approach has the potential to identify modulators of expanded repeat-containing gene expression for over 30 microsatellite expansion disorders.


Subject(s)
Drug Evaluation, Preclinical/methods , Microtubules/drug effects , Myotonic Dystrophy/genetics , RNA/genetics , Small Molecule Libraries/pharmacology , Trinucleotide Repeat Expansion/drug effects , Animals , HeLa Cells , Humans , Mice , Mice, Transgenic , Microtubules/genetics , Microtubules/metabolism , Myotonic Dystrophy/enzymology , Myotonin-Protein Kinase/genetics , Myotonin-Protein Kinase/metabolism , RNA/chemistry , RNA/metabolism
7.
Int J Mol Sci ; 19(12)2018 Dec 18.
Article in English | MEDLINE | ID: mdl-30567354

ABSTRACT

Myotonic dystrophy type 1 (DM1), the most common cause of adult-onset muscular dystrophy, is autosomal dominant, multisystemic disease with characteristic symptoms including myotonia, heart defects, cataracts and testicular atrophy. DM1 disease is being successfully modelled in Drosophila allowing to identify and validate new pathogenic mechanisms and potential therapeutic strategies. Here we provide an overview of insights gained from fruit fly DM1 models, either: (i) fundamental with particular focus on newly identified gene deregulations and their link with DM1 symptoms; or (ii) applied via genetic modifiers and drug screens to identify promising therapeutic targets.


Subject(s)
Muscle, Skeletal/physiopathology , Myotonic Dystrophy/genetics , Myotonic Dystrophy/therapy , Myotonin-Protein Kinase/genetics , Animals , Disease Models, Animal , Drosophila melanogaster/genetics , Drug Evaluation, Preclinical , Humans , Molecular Targeted Therapy , Myotonic Dystrophy/pathology , Myotonin-Protein Kinase/antagonists & inhibitors , Trinucleotide Repeat Expansion/genetics
8.
J Am Heart Assoc ; 6(4)2017 Apr 17.
Article in English | MEDLINE | ID: mdl-28416514

ABSTRACT

BACKGROUND: Cardiac arrhythmias are common causes of death in patients with myotonic dystrophy (dystrophia myotonica [DM]). Evidence shows that atrial tachyarrhythmia is an independent risk factor for sudden death; however, the relationship is unclear. METHODS AND RESULTS: Control wild-type (Mbnl1+/+; Mbnl2+/+ ) and DM mutant (Mbnl1-/-; Mbnl2+/- ) mice were generated by crossing double heterozygous knockout (Mbnl1+/-; Mbnl2+/- ) mice. In vivo electrophysiological study and optical mapping technique were performed to investigate mechanisms of ventricular tachyarrhythmias. Transmission electron microscopy scanning was performed for myocardium ultrastructural analysis. DM mutant mice were more vulnerable to anesthesia medications and program electrical pacing: 2 of 12 mice had sudden apnea and cardiac arrest during premedication of general anesthesia; 9 of the remaining 10 had atrial tachycardia and/or atrioventricular block, but none of the wild-type mice had spontaneous arrhythmias; and 9 of 10 mice had pacing-induced ventricular tachyarrhythmias, but only 1 of 14 of the wild-type mice. Optical mapping studies revealed prolonged action potential duration, slower conduction velocity, and steeper conduction velocity restitution curves in the DM mutant mice than in the wild-type group. Spatially discordant alternans was more easily inducible in DM mutant than wild-type mice. Transmission electron microscopy showed disarranged myofibrils with enlarged vacuole-occupying mitochondria in the DM mutant group. CONCLUSIONS: This DM mutant mouse model presented with clinical myofibril ultrastructural abnormality and cardiac arrhythmias, including atrial tachyarrhythmias, atrioventricular block, and ventricular tachyarrhythmias. Optical mapping studies revealed prolonged action potential duration and slow conduction velocity in the DM mice, leading to vulnerability of spatially discordant alternans and ventricular arrhythmia induction to pacing.


Subject(s)
DNA-Binding Proteins/deficiency , Myocardium/metabolism , Myofibrils/metabolism , Myotonic Dystrophy/complications , RNA-Binding Proteins/metabolism , Tachycardia, Ventricular/etiology , Ventricular Fibrillation/etiology , Voltage-Sensitive Dye Imaging , Action Potentials , Animals , Cardiac Pacing, Artificial , DNA-Binding Proteins/genetics , Disease Models, Animal , Electrophysiologic Techniques, Cardiac , Genetic Predisposition to Disease , Heart Rate , Isolated Heart Preparation , Mice, Knockout , Microscopy, Electron, Transmission , Myocardium/ultrastructure , Myofibrils/ultrastructure , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Phenotype , RNA-Binding Proteins/genetics , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/physiopathology , Time Factors , Ventricular Fibrillation/genetics , Ventricular Fibrillation/metabolism , Ventricular Fibrillation/physiopathology
9.
Trends Cardiovasc Med ; 27(1): 51-58, 2017 01.
Article in English | MEDLINE | ID: mdl-27452966

ABSTRACT

Genetic disorders that disrupt the structure and function of the cardiovascular system and the peripheral nervous system are common enough to be encountered in routine cardiovascular practice. Although often these patients are diagnosed in childhood and come to the cardiologist fully characterized, some patients with hereditary neuromuscular disease may not manifest until adulthood and will present initially to the adult cardiologist for an evaluation of an abnormal ECG, unexplained syncope, LV hypertrophy, and or a dilated cardiomyopathy of unknown cause. Cardiologists are often ill-equipped to manage these patients due to lack of training and exposure as well as the complete absence of practice guidelines to aid in the diagnosis and management of these disorders. Here, we review three key neuromuscular diseases that affect the cardiovascular system in adults (myotonic dystrophy type 1, Friedreich ataxia, and Emery-Dreifuss muscular dystrophy), with an emphasis on their clinical presentation, genetic and molecular pathogenesis, and recent important research on medical and interventional treatments. We also advocate the development of interdisciplinary cardio-neuromuscular clinics to optimize the care for these patients.


Subject(s)
Delivery of Health Care, Integrated , Heart Diseases/diagnosis , Heart Diseases/therapy , Neuromuscular Diseases/diagnosis , Neuromuscular Diseases/therapy , Patient Care Team , Adolescent , Adult , Child , Child, Preschool , Cooperative Behavior , Friedreich Ataxia/diagnosis , Friedreich Ataxia/genetics , Friedreich Ataxia/therapy , Genetic Predisposition to Disease , Genetic Testing , Heart Diseases/etiology , Heart Function Tests , Humans , Infant , Interdisciplinary Communication , Muscular Dystrophy, Emery-Dreifuss/diagnosis , Muscular Dystrophy, Emery-Dreifuss/genetics , Muscular Dystrophy, Emery-Dreifuss/therapy , Myotonic Dystrophy/diagnosis , Myotonic Dystrophy/genetics , Myotonic Dystrophy/therapy , Neuromuscular Diseases/complications , Neuromuscular Diseases/genetics , Phenotype , Predictive Value of Tests , Risk Factors , Treatment Outcome , Young Adult
10.
J Biol Chem ; 291(33): 17165-77, 2016 08 12.
Article in English | MEDLINE | ID: mdl-27298317

ABSTRACT

Myotonic dystrophy type I (DM1) is a disabling neuromuscular disease with no causal treatment available. This disease is caused by expanded CTG trinucleotide repeats in the 3' UTR of the dystrophia myotonica protein kinase gene. On the RNA level, expanded (CUG)n repeats form hairpin structures that sequester splicing factors such as muscleblind-like 1 (MBNL1). Lack of available MBNL1 leads to misregulated alternative splicing of many target pre-mRNAs, leading to the multisystemic symptoms in DM1. Many studies aiming to identify small molecules that target the (CUG)n-MBNL1 complex focused on synthetic molecules. In an effort to identify new small molecules that liberate sequestered MBNL1 from (CUG)n RNA, we focused specifically on small molecules of natural origin. Natural products remain an important source for drugs and play a significant role in providing novel leads and pharmacophores for medicinal chemistry. In a new DM1 mechanism-based biochemical assay, we screened a collection of isolated natural compounds and a library of over 2100 extracts from plants and fungal strains. HPLC-based activity profiling in combination with spectroscopic methods were used to identify the active principles in the extracts. The bioactivity of the identified compounds was investigated in a human cell model and in a mouse model of DM1. We identified several alkaloids, including the ß-carboline harmine and the isoquinoline berberine, that ameliorated certain aspects of the DM1 pathology in these models. Alkaloids as a compound class may have potential for drug discovery in other RNA-mediated diseases.


Subject(s)
3' Untranslated Regions , Alkaloids/pharmacology , DNA-Binding Proteins , Models, Biological , Myotonic Dystrophy/drug therapy , RNA-Binding Proteins , Trinucleotide Repeat Expansion , Alkaloids/chemistry , Alkaloids/isolation & purification , Alternative Splicing/drug effects , Animals , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drug Evaluation, Preclinical/methods , Humans , Mice , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Myotonic Dystrophy/pathology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
11.
Neuropathol Appl Neurobiol ; 39(4): 390-405, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22758909

ABSTRACT

AIMS: Myotonic dystrophy types 1 and 2 (DM1 and DM2) are multisystem disorders caused by similar repeat expansion mutations, with similar yet distinct clinical features. Aberrant splicing of multiple effector genes, as well as dysregulation of transcription and translation, has been suggested to underlie different aspects of the complex phenotypes in DM1 and DM2. Ca(2+) plays a central role in both muscle contraction and control of gene expression, and recent expression profiling studies have indicated major perturbations of the Ca(2+) signalling pathways in DM. Here we have further investigated the expression of genes and proteins involved in Ca(2+) metabolism in DM patients, including Ca(2+) channels and Ca(2+) binding proteins. METHODS: We used patient muscle biopsies to analyse mRNA expression and splicing of genes by microarray expression profiling and RT-PCR. We studied protein expression by immunohistochemistry and immunoblotting. RESULTS: Most of the genes studied showed mRNA up-regulation in expression profiling. When analysed by immunohistochemistry the Ca(2+) release channel ryanodine receptor was reduced in DM1 and DM2, as was calsequestrin 2, a sarcoplasmic reticulum lumen Ca(2+) storage protein. Abnormal splicing of ATP2A1 was more pronounced in DM2 than DM1. CONCLUSIONS: We observed abnormal mRNA and protein expression in DM affecting several proteins involved in Ca(2+) metabolism, with some differences between DM1 and DM2. Our protein expression studies are suggestive of a post-transcriptional defect(s) in the myotonic dystrophies.


Subject(s)
Calcium/metabolism , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Alternative Splicing , Blotting, Western , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Data Interpretation, Statistical , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Microarray Analysis , Microscopy, Confocal , Muscle, Skeletal/pathology , RNA/biosynthesis , RNA/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics
12.
J Clin Neuromuscul Dis ; 9(1): 252-5, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17989589

ABSTRACT

OBJECTIVE: To report a series of patients with adult onset myotonic dystrophy type 1 (DM1) in whom the presenting symptom was ventilatory failure. BACKGROUND: Ventilatory failure is a common complication of DM1 and may be a presenting symptom in the setting of anesthesia or surgery, but it is not known to be a heralding manifestation. METHOD: Case series. RESULTS AND DISCUSSION: Three adults developed dyspnea leading to ventilatory failure, with no cardiac or pulmonary causes identified. Case 1 required intubation for ventilator support and was sedated with propofol. There was no clinical myotonia, and electromyography (EMG) demonstrated brief runs of myotonic discharges. Examination 3 weeks later off propofol revealed percussion myotonia, and EMG evidence of long runs of myotonic discharges. Genetic testing confirmed the diagnosis of DM1. Case 2 had cataracts and ptosis but no known diagnosis of DM and no previous neurological impairments. Case 3 was previously neurologically asymptomatic but her son had congenital DM1. The diagnosis was confirmed by EMG in cases 2 and 3, and both patients were managed with bilevel ventilation (BIPAP). CONCLUSION: Myotonic dystrophy type 1 should be considered in the differential diagnosis of acute ventilatory failure in adults.


Subject(s)
Myotonic Dystrophy/complications , Respiratory Insufficiency/etiology , Adult , Electromyography , Family Health , Female , Humans , Middle Aged , Myotonic Dystrophy/classification , Myotonic Dystrophy/genetics , Neurologic Examination/methods , Plant Extracts/genetics , Respiratory Insufficiency/diagnosis
13.
Biochim Biophys Acta ; 1772(11-12): 1183-91, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17950578

ABSTRACT

Myotonic dystrophy (DM1) is a dominant autosomal multisystemic disorder caused by the expansion of an unstable CTG trinucleotide repeat in the 3' untranslated region of the DMPK gene. Nuclear accumulation of the enlarged CUG-containing DMPK transcripts has a deleterious effect on the regulation of alternative splicing of some RNAs and has a central role in causing the symptoms of DM1. In particular, Insulin Receptor (IR) mRNA splicing defects have been observed in the muscle of DM1 patients. In this study, we have investigated IR splicing in insulin-responsive tissues (i.e. skeletal muscles, adipose tissue, liver) and pancreas and we have studied glucose metabolism in mice carrying the human genomic DM1 region with expanded (>350 CTG) or normal (20 CTG) repeats and in wild-type mice. Mice carrying DM1 expansions displayed a tissue- and age-dependent abnormal regulation of IR mRNA splicing in all the tissues that we investigated. Furthermore, these mice showed a basal hyperglycemia and glucose intolerance which disappeared with age. Our findings show that deregulation of IR splicing due to the DM1 mutation can occur in different mouse tissues, suggesting that CTG repeat expansions might also result in IR misplicing not only in muscles but also in other tissues in DM1 patients.


Subject(s)
Myotonic Dystrophy/genetics , Receptor, Insulin/genetics , Trinucleotide Repeat Expansion/genetics , Aging , Alternative Splicing/genetics , Animals , Gene Expression Profiling , Glucose/metabolism , Glucose Tolerance Test , Humans , Hypothalamus/metabolism , Insulin/metabolism , Insulin Secretion , Mice , Mice, Transgenic , Mutant Proteins/metabolism , Myotonin-Protein Kinase , Organ Specificity , Pancreas/enzymology , Pancreas/pathology , Protein Isoforms/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Insulin/metabolism , Transgenes
14.
Fam Process ; 44(1): 25-44, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15807076

ABSTRACT

The evolution of genomic science and its effect on medicine and health care offer opportunities for family therapists to participate in the comprehensive care of patients and families with genetic disorders. This article provides an overview of what we now know about the psychological and interpersonal experience of patients and families facing some of these illnesses. Case examples illustrate the process of decision-making about testing and treatment, and the importance of understanding developmental issues and transgenerational family dynamics in any related psychotherapy. Challenging emotional issues include managing anger, ambivalence, and guilt; challenging interpersonal issues include dealing with differing coping and communication styles, decisions about disclosure and secrets, and conflict resolution. Family-oriented interventions include individual, couple, and family therapy, and psychoeducational groups. Recommendations are made for family therapists to participate as part of the genetic healthcare team.


Subject(s)
Family Relations , Family Therapy , Genetic Counseling/psychology , Genetic Diseases, Inborn/psychology , Genetic Testing/psychology , Parent-Child Relations , Adolescent , Adult , Anger , Breast Neoplasms/genetics , Breast Neoplasms/psychology , Child , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/psychology , Emotions , Female , Genetic Diseases, Inborn/genetics , Genetic Predisposition to Disease , Guilt , Humans , Huntington Disease/genetics , Huntington Disease/psychology , Intergenerational Relations , Male , Myotonic Dystrophy/genetics , Myotonic Dystrophy/psychology , Patient Education as Topic , Pedigree
15.
Neurology ; 63(12): 2371-5, 2004 Dec 28.
Article in English | MEDLINE | ID: mdl-15623702

ABSTRACT

BACKGROUND: Muscle fiber degeneration and myotonic discharges are the hallmarks of myotonic dystrophy (DM). The molecular basis for the myotonia was recently tied to abnormal splicing of the chloride channel (ClC-1) pre-mRNA, often resulting in UAG premature termination, which leads to decreased channel protein and therefore a reduced resting chloride conductance. METHODS: The authors assessed the functional properties of two commonly occurring DM mRNA splice variants by expression in oocytes. RESULTS: Neither splice variant coded for a functional Cl- channel. Co-injection of alternative splice variants with wild-type ClC-1 cRNA reduced the current density and accelerated channel closure upon repolarization of the membrane. CONCLUSIONS: These data show that the aberrantly spliced chloride channel message exerts a dominant negative effect that may contribute to the development of myotonia.


Subject(s)
Alternative Splicing , Chloride Channels/genetics , Chlorides/metabolism , Codon, Nonsense , Myotonic Dystrophy/genetics , RNA Precursors/genetics , RNA, Messenger/genetics , Animals , DNA, Complementary/genetics , Exons/genetics , Female , Frameshift Mutation , Humans , Ion Transport/genetics , Membrane Potentials , Microinjections , Models, Molecular , Oocytes , Patch-Clamp Techniques , Protein Conformation , RNA/genetics , Xenopus laevis
16.
Fisioterapia (Madr., Ed. impr.) ; 25(2): 110-120, abr. 2003. ilus, tab
Article in Es | IBECS | ID: ibc-24440

ABSTRACT

En los últimos años han llegado a nuestra Unidad de Pediatría cinco casos clínicos diagnosticados de Síndrome de Steinert neonatal. Ante la aparente ausencia de bibliografía con que nos hemos encontrado, nos parece interesante la elaboración de un protocolo de tratamiento fisioterápico que se ajuste a los resultados más satisfactorios de las técnicas que hemos empleado, así como compartir información sobre los casos y su evolución durante sus primeros tres años de vida (AU)


Subject(s)
Humans , Infant, Newborn , Myotonic Dystrophy/rehabilitation , Clinical Protocols , Physical Therapy Specialty/methods , Myotonic Dystrophy/genetics , Clinical Evolution , Bottle Feeding , Facial Muscles/physiology , Equinus Deformity/rehabilitation , Massage , Respiration Disorders/rehabilitation
17.
Biochem Biophys Res Commun ; 281(4): 835-41, 2001 Mar 09.
Article in English | MEDLINE | ID: mdl-11237735

ABSTRACT

Ciliary function is essential for normal cellular activity in all species from simple protozoa upwards. In humans, ciliary dysmotility or complete immobility have been identified in autosomal recessive multisystemic diseases characterized by recurrent respiratory tract infections and male subfertility due to impaired sperm mobility. Linkage to human chromosome 19q13.3 has been published for some families but no candidate genes have been identified. We report the first identification of a mammalian homolog of a radial spokehead-like protein, with high homology to proteins of sea urchins and the protozoan Chlamydomonas reinhardtii, at the myotonic dystrophy-1 locus (chromosome19q13.3). In the lower organisms, these proteins are important in normal ciliary or flagellar action, including that of sea urchin spermatozoa. Expression of the mammalian homolog was detected in the adult testis. We suggest that this gene, which we have called Radial Spokehead-Like 1 (RSHL1), is a candidate gene for familial primary ciliary dyskinesia.


Subject(s)
Ciliary Motility Disorders/genetics , Iron-Binding Proteins , Myotonic Dystrophy/genetics , Proteins/genetics , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Chromosomes, Human, Pair 19/genetics , DNA, Complementary/chemistry , DNA, Complementary/genetics , Female , Gene Expression , Humans , Male , Mice , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Tissue Distribution
20.
J Clin Endocrinol Metab ; 41(5): 833-40, 1975 Nov.
Article in English | MEDLINE | ID: mdl-1184720

ABSTRACT

Gonadal function was studied in three post-pubertal siblings (two male and one female) and one unrelated male patient with myotonic dystrophy. The diagnosis was confirmed in all cases by electromyography and muscle biopsy. Basal levels of plasma immunoreactive LH, FSH, testosterone, and estradiol were measured. Hypothalamic, pituitary, and gonadal reserve and responsiveness were evaluated by clomiphene, LHRH, and HCG tests. Histologic examination of gonadal biopsies was also performed. The results showed that gonadal failure present in the four patients had different characteristics. In the same family, hypothalamic amenorrhea was observed in the female patient, and hypothalamic eunuchoidism and hypergonadotropic hypogonadism with marked tubular and leydig cells failure in the male patients. The non-related male patient had hypergonadotropic hypogonadism with tubular failure but with a compensatory leydig-cell hyperplasia. These data are interpreted as demonstrating different expressivity of the hypogonadism associated with the same inherited muscle disease.


Subject(s)
Hypogonadism/physiopathology , Hypothalamus/physiopathology , Myotonic Dystrophy/physiopathology , Pituitary Gland/physiopathology , Testis/physiopathology , Adult , Clomiphene/pharmacology , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Genes, Dominant , Humans , Hypogonadism/blood , Hypogonadism/genetics , Luteinizing Hormone/blood , Male , Myotonic Dystrophy/blood , Myotonic Dystrophy/genetics , Pedigree , Progesterone/pharmacology , Testis/pathology , Testosterone/blood
SELECTION OF CITATIONS
SEARCH DETAIL