Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
1.
BMJ Case Rep ; 17(6)2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38866581

ABSTRACT

Nemaline rod myopathy is an extremely rare muscle disease responsible for hypotonia and poor muscle strength in infants. The disease has variable phenotypic presentations across different ages, ranging from neonatal to the adult onset and from severe to asymptomatic varieties. Clinical features, muscle biopsy and genetic testing help in diagnosis. The histopathological examination shows the presence of rod-like structures or nemaline bodies in muscles. Management remains mainly supportive, and currently, there is no available curative treatment. This case report describes an infant presenting with gross hypotonia, poor handling of secretions and multiple extubation failures who was diagnosed by clinical exome sequencing. The patient harboured compound heterozygous variants in the NEB gene suggestive of nemaline rod myopathy. The newborn showed significant improvement in muscle strength after he was started on dietary L-tyrosine supplementation. This case highlights the emerging role of L-tyrosine in the supportive care of infants with nemaline rod myopathy.


Subject(s)
Myopathies, Nemaline , Tyrosine , Humans , Myopathies, Nemaline/genetics , Myopathies, Nemaline/drug therapy , Male , Tyrosine/therapeutic use , Infant, Newborn , Infant , Muscle Hypotonia/drug therapy , Muscle Proteins/genetics , Muscle, Skeletal/pathology , Treatment Outcome
2.
Horm Res Paediatr ; 97(2): 148-156, 2024.
Article in English | MEDLINE | ID: mdl-37343528

ABSTRACT

INTRODUCTION: Schaaf-Yang syndrome (SYS) is a rare neurodevelopmental disorder caused by truncating mutations of the MAGEL2 gene, located in the Prader-Willi syndrome (PWS) region. PWS and SYS have phenotypic overlap. Patients with SYS are often treated with growth hormone (GH), but evidence for the effectiveness of the treatment in patients with SYS is limited. METHODS: This study describes 7 children with SYS. We studied their phenotype, genotype, and the effect of GH treatment on height and body mass index (BMI) during 4 years and on body composition during 1 year. RESULTS: All patients had a normal birth weight. Most patients had hypotonia and feeding difficulties after birth (86%). Full-scale IQ ranged from <50 to 92. All patients above the age of 2 years had psycho-behavioral problems. There were no apparent correlations between the phenotype and the location of the defect in the MAGEL2 gene. Mean (95% CI) height SDS increased significantly from -1.74 (-3.55; 0.07) at start to -0.05 (-1.87; 1.77) after 4 years of GH treatment. Mean (95% CI) BMI SDS decreased significantly from 2.01 (1.02; 3.00) to 1.22 (0.18; 2.26) after 6 months and remained the same during the rest of the follow-up. Fat mass percentage SDS decreased and lean body mass did not change during 1 year of treatment in 3 patients. CONCLUSION: Patients presented with a phenotype of hypotonia, respiratory insufficiency, and feeding difficulties after birth, endocrine disorders, intellectual disability, and behavioral problems. Treatment with GH significantly improved height SDS and BMI over the course of 4 years.


Subject(s)
Chromosome Disorders , Developmental Disabilities , Facies , Human Growth Hormone , Hypopituitarism , Imprinting Disorders , Child , Child, Preschool , Humans , Human Growth Hormone/therapeutic use , Human Growth Hormone/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intrinsically Disordered Proteins/genetics , Muscle Hypotonia/drug therapy , Muscle Hypotonia/genetics , Phenotype , Prader-Willi Syndrome/drug therapy , Prader-Willi Syndrome/genetics
3.
J Clin Res Pediatr Endocrinol ; 16(1): 116-122, 2024 03 11.
Article in English | MEDLINE | ID: mdl-38054413

ABSTRACT

Monocarboxylate transporter 8 (MCT8) deficiency is a rare genetic disorder characterized by peripheral thyrotoxicosis and severe cognitive and motor disability due to cerebral hypothyroidism. 3,3',5-triiodothyroacetic acid (Triac) was shown to improve peripheral thyrotoxicosis but data on neurodevelopmental outcome are scarce. We present a case of MCT8 deficiency and the experience with Triac focusing on change in neurodevelopmental and peripheral features. A five-month-old boy was referred because of feeding difficulty, central hypotonia and global developmental delay. Despite six months of physiotherapy, physical developmental milestones did not improve, and distal muscle tone was increased. A hemizygous pathogenic variant in SLC16A2 was found and MCT8 deficiency was confirmed at 19-months. Thyroid stimulating hormone was 2.83 mIU/mL, free thyroxine 6.24 pmol/L (N=12-22) and free triiodothyronine (FT3) 15.65pmol/L (N=3.1-6.8). He had tachycardia, blood pressure and transaminases were elevated. Triac was started at 21-months. Two weeks after treatment, FT3 dramatically decreased, steady normal serum FT3 was achieved at 28-months. Assessment of neurodevelopmental milestones and signs of hyperthyroidism were evaluated at baseline, 6 months and 12 months after treatment. Signs of hyperthyroidism were improved by 6 months. Developmental composite scores of Bayley Scales of Infant Developmental 3rd Edition remained the same but important developmental milestones (head control, recognition of caregiver, response to his name) were attained, regression in the attained milestones were not observed. Initial dose, management protocol for Triac and research into its efficacy on neurodevelopmental signs in MCT8 deficiency are progressing. This case presents evidence that Triac may resolve peripheral thyrotoxicosis successfully and may slow neurodevelopmental regression, while some developmental milestones were achieved after one year of treatment.


Subject(s)
Disabled Persons , Hyperthyroidism , Mental Retardation, X-Linked , Motor Disorders , Symporters , Thyrotoxicosis , Triiodothyronine/analogs & derivatives , Male , Infant , Humans , Muscle Hypotonia/diagnosis , Muscle Hypotonia/drug therapy , Muscle Hypotonia/genetics , Mental Retardation, X-Linked/diagnosis , Muscular Atrophy/diagnosis , Muscular Atrophy/drug therapy , Muscular Atrophy/genetics , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/therapeutic use , Symporters/genetics , Symporters/therapeutic use
4.
Bone ; 172: 116778, 2023 07.
Article in English | MEDLINE | ID: mdl-37088336

ABSTRACT

Tissue non-specific alkaline phosphatase (TNSALP) is an enzyme that is tethered to the cell membrane by glycosylphosphatidylinositol (GPI) and converts inorganic pyrophosphate to inorganic phosphate. Inorganic phosphate combines with calcium to form hydroxyapatite, the main mineral in the skeleton. When TNSALP is defective, conversion of inorganic pyrophosphate to inorganic phosphate is impaired and the skeleton is at risk of under-mineralization. Phosphatidylinositol glycan anchor biosynthesis class N (PIGN) is one of more than 20 genes in the GPI-biosynthesis family. Pathogenic variants in PIGN have been identified in multiple congenital anomalies-hypotonia-seizures syndrome (OMIM 614080), although a metabolic bone disease or skeletal fragility phenotype has not been reported. We describe a female child with multiple congenital anomalies-hypotonia-seizures syndrome due to a compound heterozygous pathogenic variant in PIGN who sustained a low-trauma distal femur fracture at age 7.4 years. We hypothesized that the GPI synthesis defect may result in metabolic bone disease from inadequate anchoring of TNSALP in bone and initiated asfotase alfa, a human bone-targeted recombinant TNSALP-Fc-deca-aspartate peptide, as it could bypass the PIGN genetic defect that possibly caused her skeletal fragility. Asfotase alfa was begun at 8.5 years. Baseline X-rays revealed mild rachitic findings of wrists and knees, which resolved by 5 months of treatment. Bone mineral density (BMD) assessed by dual-energy X-ray absorptiometry (DXA) showed mild improvement in spine, hip and total body less head after 16 months of treatment, while radius declined. She sustained additional low trauma fractures at right tibia and left humeral neck at 11 and 15 months into treatment, which healed quickly. Calcium, phosphorus, and parathyroid hormone levels have remained within the normal range over the 18 months of treatment. For adverse effect, she experienced a rash and discomfort in the first week of treatment which resolved with ibuprofen and diphenhydramine. She also developed subcutaneous fat atrophy. Overall, in this child with a compound pathogenic variant in PIGN, off-label use of asfotase alfa has been generally well tolerated with minimal side effects and resolution of rickets, but she continues to remain skeletally fragile.


Subject(s)
Bone Diseases, Metabolic , Calcinosis , Hypophosphatasia , Osteoporotic Fractures , Humans , Child , Female , Alkaline Phosphatase/therapeutic use , Hypophosphatasia/drug therapy , Hypophosphatasia/genetics , Diphosphates , Calcium/therapeutic use , Fracture Healing , Muscle Hypotonia/drug therapy , Bone and Bones , Bone Diseases, Metabolic/drug therapy , Calcinosis/drug therapy , Calcium, Dietary , Osteoporotic Fractures/drug therapy , Seizures/drug therapy
5.
Am J Med Genet A ; 188(6): 1831-1835, 2022 06.
Article in English | MEDLINE | ID: mdl-35156754

ABSTRACT

This case report describes a patient with early-onset cobalamin C deficiency who was started on treatment with high-dose parenteral hydroxocobalamin after diagnosis at 13 days of life. Prior to diagnosis, initial presenting symptoms included poor feeding, lethargy, apneic episodes, hypothermia, and hypotonia; these symptoms resolved after initiation of medication. Methylmalonic acid and homocysteine levels were trended and significantly improved with treatment. She was maintained on 2 mg/kg/day dosing of hydroxocobalamin. No adverse effects to treatment were observed. At the time of this report, the patient was 19 months of age; she had not manifested common findings of early-onset cobalamin C deficiency, including microcephaly, poor feeding, growth abnormalities, hypotonia, seizures, maculopathy, or neurodevelopmental delay. This report suggests that early initiation of high-dose hydroxocobalamin is safe and effective.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Vitamin B 12 Deficiency , Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/drug therapy , Female , Humans , Hydroxocobalamin/therapeutic use , Infant, Newborn , Methylmalonic Acid , Muscle Hypotonia/drug therapy , Vitamin B 12 Deficiency/diagnosis , Vitamin B 12 Deficiency/drug therapy
6.
J Clin Endocrinol Metab ; 107(3): e1136-e1147, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34679181

ABSTRACT

CONTEXT: Patients with mutations in thyroid hormone transporter MCT8 have developmental delay and chronic thyrotoxicosis associated with being underweight and having cardiovascular dysfunction. OBJECTIVE: Our previous trial showed improvement of key clinical and biochemical features during 1-year treatment with the T3 analogue Triac, but long-term follow-up data are needed. METHODS: In this real-life retrospective cohort study, we investigated the efficacy of Triac in MCT8-deficient patients in 33 sites. The primary endpoint was change in serum T3 concentrations from baseline to last available measurement. Secondary endpoints were changes in other thyroid parameters, anthropometric parameters, heart rate, and biochemical markers of thyroid hormone action. RESULTS: From October 15, 2014 to January 1, 2021, 67 patients (median baseline age 4.6 years; range, 0.5-66) were treated up to 6 years (median 2.2 years; range, 0.2-6.2). Mean T3 concentrations decreased from 4.58 (SD 1.11) to 1.66 (0.69) nmol/L (mean decrease 2.92 nmol/L; 95% CI, 2.61-3.23; P < 0.0001; target 1.4-2.5 nmol/L). Body-weight-for-age exceeded that of untreated historical controls (mean difference 0.72 SD; 95% CI, 0.36-1.09; P = 0.0002). Heart-rate-for-age decreased (mean difference 0.64 SD; 95% CI, 0.29-0.98; P = 0.0005). SHBG concentrations decreased from 245 (99) to 209 (92) nmol/L (mean decrease 36 nmol/L; 95% CI, 16-57; P = 0.0008). Mean creatinine concentrations increased from 32 (11) to 39 (13) µmol/L (mean increase 7 µmol/L; 95% CI, 6-9; P < 0.0001). Mean creatine kinase concentrations did not significantly change. No drug-related severe adverse events were reported. CONCLUSIONS: Key features were sustainably alleviated in patients with MCT8 deficiency across all ages, highlighting the real-life potential of Triac for MCT8 deficiency.


Subject(s)
Mental Retardation, X-Linked/drug therapy , Monocarboxylic Acid Transporters/deficiency , Muscle Hypotonia/drug therapy , Muscular Atrophy/drug therapy , Symporters/deficiency , Triiodothyronine/analogs & derivatives , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Infant , Male , Mental Retardation, X-Linked/blood , Mental Retardation, X-Linked/genetics , Middle Aged , Monocarboxylic Acid Transporters/genetics , Muscle Hypotonia/blood , Muscle Hypotonia/genetics , Muscular Atrophy/blood , Muscular Atrophy/genetics , Mutation , Retrospective Studies , Symporters/genetics , Treatment Outcome , Triiodothyronine/administration & dosage , Triiodothyronine/adverse effects , Triiodothyronine/blood , Young Adult
7.
J Coll Physicians Surg Pak ; 32(12): SS134-SS136, 2022 12.
Article in English | MEDLINE | ID: mdl-36597316

ABSTRACT

Pompe disease, also known as Glycogen Storage Disease Type II, is a rare disorder of glucose metabolism caused by congenital acid alpha-glucosidase (GAA) deficiency. A large amount of glycogen accumulates in the lysosomes, causing these to swell and rupture. Its incidence is about 1 in 40,000 to 1 in 50,000 newborns. The main features are hypotonia and cardiomyopathy. Only a few clinical cases of Pompe disease have been reported, and appendicular torsion has rarely been observed. Herein, we report a case of Pompe disease combined with appendicular torsion, both of which were diagnosed on autopsy pathology. The clinical diagnosis of this disease is difficult in developing countries, and it is mostly misdiagnosed as other types of heart disease. Once the clinical symptoms worsen, most of them die within a short period. Therefore, screening for neonatal genetic metabolic diseases for early diagnosis and treatment should be carried out. Key Words: Glycogen storage disease type II, Metabolic disease, Enzyme replacement therapy, Neonatal screening.


Subject(s)
Cardiomyopathies , Glycogen Storage Disease Type II , Infant, Newborn, Diseases , Humans , Infant, Newborn , Glycogen Storage Disease Type II/complications , Glycogen Storage Disease Type II/diagnosis , Glycogen Storage Disease Type II/genetics , alpha-Glucosidases/genetics , alpha-Glucosidases/metabolism , alpha-Glucosidases/therapeutic use , Muscle Hypotonia/drug therapy , Muscle Hypotonia/metabolism , Cardiomyopathies/drug therapy , Lysosomes/metabolism , Lysosomes/pathology
8.
Curr Drug Discov Technol ; 18(1): 105-112, 2021.
Article in English | MEDLINE | ID: mdl-31774046

ABSTRACT

AIMS: To identify natural inhibitors against MCT8 for Allan-Herndon-Dudley Syndrome. BACKGROUND: Monocarboxylate Transporter 8 (MCT8) is a Thyroid Hormone (TH) transporter which is highly expressed in the liver and brain. Mutations in the MCT8 gene (SLC16A2) cause a syndrome of psychomotor retardation in humans, known as Allan-Herndon-Dudley syndrome (AHDS). Currently, no treatment is available for AHDS. Therefore, there is a need to discover new inhibitors of MCT8 for treating AHDS. OBJECTIVE: Considering the importance of natural compounds in drug discovery, this study aimed to identify potential natural inhibitors against MCT8. METHODS: As Protein-ligand interactions play a key role in structure based drug design, this study screened 24 natural kinase inhibitors and investigated their binding affinity against MCT8 by using molecular docking. The modelled 3D structure of MCT8 docked with 24 compounds using PyRX through Autodock Vina. Drug-likeness studies were made using Swiss ADME and Lipinski's rule of five was performed. Triac, desipramine and silychristin were used as the positive controls. Binding energies of the selected compounds were compared with that of positive controls. RESULT: The results showed that emodin exhibited best binding energy of -8.6 kcal/mol followed by helenaquinol, cercosporamide and resveratrol. Moreover, it was observed that emodin and helenaquinol exhibit higher binding energy than the positive controls. Cercosporamide and resveratrol exhibited higher binding energy than triac and desipramine and showed the binding energy similar to silychristin. CONCLUSION: This study reveals that these compounds could be promising candidates for further evaluation for AHDS prevention.


Subject(s)
Benzofurans/pharmacology , Emodin/pharmacology , Mental Retardation, X-Linked , Monocarboxylic Acid Transporters , Muscle Hypotonia , Muscular Atrophy , Resveratrol/pharmacology , Symporters , Biological Transport/drug effects , Drug Discovery/methods , Humans , Mental Retardation, X-Linked/drug therapy , Mental Retardation, X-Linked/genetics , Molecular Docking Simulation/methods , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/genetics , Muscle Hypotonia/drug therapy , Muscle Hypotonia/genetics , Muscular Atrophy/drug therapy , Muscular Atrophy/genetics , Mutation , Phytochemicals/pharmacology , Protein Kinase Inhibitors/pharmacology , Symporters/antagonists & inhibitors , Symporters/genetics , Thyroid Hormones/metabolism
9.
Am J Med Genet A ; 185(1): 256-260, 2021 01.
Article in English | MEDLINE | ID: mdl-33098379

ABSTRACT

Early-onset severe spinocerebellar ataxia 42 with neurodevelopmental deficits (SCA42ND, MIM#604065) is an ultrarare autosomal dominant syndrome related to de novo CACNA1G gain-of-function pathogenic variants. All patients with SCA42ND show cerebellar atrophy and/or hypoplasia on neuroimaging and share common features such as dysmorphic features, global developmental delay, and axial hypotonia, all manifesting within the first year of life. To date, only 10 patients with SCA42ND have been reported with functionally confirmed gain-of-function variants, bearing either of two recurrent pathogenic variants. We describe a girl with congenital ataxia, without epilepsy, and a de novo p.Ala961Thr pathogenic variant in CACNA1G. We review the published subjects with the aim of better characterizing the dysmorphic features that may be crucial for clinical recognition of SCA42ND. Cerebellar atrophy, together with digital anomalies, particularly broad thumbs and/or halluces, should lead to clinical suspicion of this disease. We describe the first pharmacological attempt to treat a patient with SCA42ND using zonisamide, an antiepileptic drug with T-type channel blocker activity, in an off-label indication using an itemized study protocol. No efficacy was observed at the dose tested. However, without pharmacological treatment, she showed a positive evolution in neurodevelopment during the follow-up.


Subject(s)
Calcium Channels, T-Type/genetics , Epilepsy/genetics , Muscle Hypotonia/genetics , Spinocerebellar Ataxias/genetics , Age of Onset , Alleles , Child, Preschool , Epilepsy/complications , Epilepsy/diagnostic imaging , Epilepsy/drug therapy , Female , Gain of Function Mutation/genetics , Humans , Infant , Male , Muscle Hypotonia/complications , Muscle Hypotonia/diagnostic imaging , Muscle Hypotonia/drug therapy , Mutation , Pedigree , Phenotype , Spinocerebellar Ataxias/complications , Spinocerebellar Ataxias/diagnostic imaging , Spinocerebellar Ataxias/drug therapy , Zonisamide/administration & dosage
10.
Thyroid ; 31(5): 713-720, 2021 05.
Article in English | MEDLINE | ID: mdl-32746752

ABSTRACT

Background: Mutations of the thyroid hormone (TH)-specific cell membrane transporter, monocarboxylate transporter 8 (MCT8), produce an X-chromosome-linked syndrome of TH deficiency in the brain and excess in peripheral tissues. The clinical consequences include brain hypothyroidism causing severe psychoneuromotor abnormalities (no speech, truncal hypotonia, and spastic quadriplegia) and hypermetabolism (poor weight gain, tachycardia, and increased metabolism, associated with high serum levels of the active TH, T3). Treatment in infancy and childhood with TH analogues that reduce serum triiodothyronine (T3) corrects hypermetabolism, but has no effect on the psychoneuromotor deficits. Studies of brain from a 30-week-old MCT8-deficient embryo indicated that brain abnormalities were already present during fetal life. Methods: A carrier woman with an affected male child (MCT8 A252fs268*), pregnant with a second affected male embryo, elected to carry the pregnancy to term. We treated the fetus with weekly 500 µg intra-amniotic instillation of levothyroxine (LT4) from 18 weeks of gestation until birth at 35 weeks. Thyroxine (T4), T3, and thyrotropin (TSH) were measured in the amniotic fluid and maternal serum. Treatment after birth was continued with LT4 and propylthiouracil. Follow-up included brain magnetic resonance imaging (MRI) and neurodevelopmental evaluation, both compared with the untreated brother. Results: During intrauterine life, T4 and T3 in the amniotic fluid were maintained above threefold to twofold the baseline and TSH was suppressed by 80%, while maternal serum levels remained unchanged. At birth, the infant serum T4 was 14.5 µg/dL and TSH <0.01 mU/L compared with the average in untreated MCT8-deficient infants of 5.1 µg/ and >8 mU/L, respectively. MRI at six months of age showed near-normal brain myelination compared with much reduced in the untreated brother. Neurodevelopmental assessment showed developmental quotients in receptive language and problem-solving, and gross motor and fine motor function ranged from 12 to 25 at 31 months in the treated boy and from 1 to 7 at 58 months in the untreated brother. Conclusions: This is the first demonstration that prenatal treatment improved the neuromotor and neurocognitive function in MCT8 deficiency. Earlier treatment with TH analogues that concentrate in the fetus when given to the mother may further rescue the phenotype.


Subject(s)
Antithyroid Agents/therapeutic use , Fetal Therapies/methods , Mental Retardation, X-Linked/drug therapy , Muscle Hypotonia/drug therapy , Muscular Atrophy/drug therapy , Propylthiouracil/therapeutic use , Thyroxine/therapeutic use , Adult , Amniotic Fluid , Brain/diagnostic imaging , Case-Control Studies , Female , Humans , Magnetic Resonance Imaging , Male , Mental Retardation, X-Linked/diagnostic imaging , Mental Retardation, X-Linked/physiopathology , Monocarboxylic Acid Transporters/genetics , Muscle Hypotonia/diagnostic imaging , Muscle Hypotonia/physiopathology , Muscular Atrophy/diagnostic imaging , Muscular Atrophy/physiopathology , Pregnancy , Symporters/genetics , Thyrotropin/metabolism , Thyroxine/metabolism , Triiodothyronine/metabolism
11.
Yonsei Med J ; 61(11): 981-985, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33107243

ABSTRACT

3-M syndrome is a rare autosomal recessive growth disorder characterized by severe growth retardation, low birth weight, characteristic facial features, and skeletal anomalies, for which three causative genes (CUL7, OBSL1, and CCDC8) have been identified. We herein report two Korean siblings with 3-M syndrome caused by two novel OBSL1 mutations, and describe the effect of a combined treatment with growth hormone (GH) and a gonadotropin-releasing hormone (GnRH) agonist. A 7-year-old girl with short stature (-3.37 standard deviation score, SDS) and breast budding presented with subtle dysmorphic features, including macrocephaly, frontal bossing, a triangular face, prominent philtrum, full lips, a short neck, and fifth-finger clinodactyly. GnRH stimulation test revealed a pubertal pattern and advanced bone age of 8 years and 10 months. Her older sister, aged 10 years and 9 months, had experienced an early menarche, and had an advanced bone age (13.5 years) and predicted adult height of 142 cm (-4.04 SDS). Targeted exome sequencing identified that the siblings had two heteroallelic mutations in OBSL1. Both siblings underwent a combination therapy with GH and a GnRH agonist. A height gain was noted in both siblings even after short-term treatment. To fully elucidate the effects of the combined therapy, a larger cohort should be analyzed following a longer treatment period. However, such an analysis would be challenging due to the rarity of this disease.


Subject(s)
Cytoskeletal Proteins/genetics , Dwarfism/drug therapy , Gonadotropin-Releasing Hormone/therapeutic use , Human Growth Hormone/therapeutic use , Muscle Hypotonia/drug therapy , Puberty, Precocious/genetics , Spine/abnormalities , Child , Dwarfism/diagnosis , Female , Gonadotropin-Releasing Hormone/agonists , Humans , Muscle Hypotonia/diagnosis , Mutation , Republic of Korea , Siblings , Treatment Outcome , Exome Sequencing
12.
J Pediatr Endocrinol Metab ; 33(12): 1609-1612, 2020 Dec 16.
Article in English | MEDLINE | ID: mdl-32924381

ABSTRACT

OBJECTIVE: 3-M syndrome is characterized by severe short stature, syndromic features, and characteristic radiographic findings. Growth hormone (GH) has been used with variable success. Recombinant human insulin like growth factor-1 (rhIGF-1) has never been utilized. CASE PRESENTATION: We describe a child with severe growth retardation, macrocephaly, and skeletal abnormalities with evidence of GH insensitivity subsequently treated with rhIGF-1. He developed morbid obesity and comorbidities including voracious appetite, acanthosis nigricans, tonsillar hypertrophy, and severe obstructive sleep apnea with minimal height improvement. Genetic testing done at 11.5 years revealed a compound heterozygous mutation (c.2112G>A(p.W704X) and c.2559delC) in the CUL7 gene consistent with 3-M syndrome-1. rhIGF-1 therapy was discontinued. CONCLUSIONS: This case highlights the novel use of rhIGF-1 therapy on a child with 3-M syndrome-1 with minimal height benefit but accelerated weight gain and serves as a reminder of the importance of re-evaluating therapy efficacy and side effect profile.


Subject(s)
Cullin Proteins/genetics , Dwarfism/drug therapy , Growth Substances/therapeutic use , Insulin-Like Growth Factor I/therapeutic use , Muscle Hypotonia/drug therapy , Mutation , Recombinant Proteins/therapeutic use , Spine/abnormalities , Child, Preschool , Dwarfism/etiology , Dwarfism/pathology , Humans , Male , Muscle Hypotonia/etiology , Muscle Hypotonia/pathology , Prognosis , Spine/pathology
13.
Adv Genet ; 105: 137-174, 2020.
Article in English | MEDLINE | ID: mdl-32560786

ABSTRACT

Potassium channels are a heterogeneous group of membrane-bound proteins, whose functions support a diverse range of biological processes. Genetic disorders arising from mutations in potassium channels are classically recognized by symptoms arising from acute channel dysfunction, such as periodic paralysis, ataxia, seizures, or cardiac conduction abnormalities, often in a patient with otherwise normal examination findings. In this chapter, we review a distinct subgroup of rare potassium channelopathies whose presentations are instead suggestive of a developmental disorder, with features including intellectual disability, craniofacial dysmorphism or other physical anomalies. Known conditions within this subgroup are: Andersen-Tawil syndrome, Birk-Barel syndrome, Cantú syndrome, Keppen-Lubinsky syndrome, Temple-Baraitser syndrome, Zimmerman-Laband syndrome and a very similar disorder called Bauer-Tartaglia or FHEIG syndrome. Ion channelopathies are unlikely to be routinely considered in the differential diagnosis of children presenting with developmental concerns, and so detailed description and photographs of the clinical phenotype are provided to aid recognition. For several of these disorders, functional characterization of the genetic mutations responsible has led to identification of candidate therapies, including drugs already commonly used for other indications, which adds further impetus to their prompt recognition. Together, these cases illustrate the potential for mechanistic insights gained from genetic diagnosis to drive translational work toward targeted, disease-modifying therapies for rare disorders.


Subject(s)
Abnormalities, Multiple/genetics , Andersen Syndrome/genetics , Cardiomegaly/genetics , Channelopathies/genetics , Craniofacial Abnormalities/genetics , Fibromatosis, Gingival/genetics , Hallux/abnormalities , Hand Deformities, Congenital/genetics , Hypertrichosis/genetics , Intellectual Disability/genetics , Muscle Hypotonia/genetics , Nails, Malformed/genetics , Osteochondrodysplasias/genetics , Potassium Channels/genetics , Thumb/abnormalities , Abnormalities, Multiple/drug therapy , Abnormalities, Multiple/pathology , Abnormalities, Multiple/physiopathology , Andersen Syndrome/drug therapy , Andersen Syndrome/pathology , Andersen Syndrome/physiopathology , Cardiomegaly/drug therapy , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Channelopathies/drug therapy , Channelopathies/metabolism , Channelopathies/physiopathology , Child , Craniofacial Abnormalities/drug therapy , Craniofacial Abnormalities/pathology , Craniofacial Abnormalities/physiopathology , Fibromatosis, Gingival/drug therapy , Fibromatosis, Gingival/pathology , Fibromatosis, Gingival/physiopathology , Hallux/pathology , Hallux/physiopathology , Hand Deformities, Congenital/drug therapy , Hand Deformities, Congenital/pathology , Hand Deformities, Congenital/physiopathology , Humans , Hypertrichosis/drug therapy , Hypertrichosis/pathology , Hypertrichosis/physiopathology , Intellectual Disability/drug therapy , Intellectual Disability/pathology , Intellectual Disability/physiopathology , Muscle Hypotonia/drug therapy , Muscle Hypotonia/pathology , Muscle Hypotonia/physiopathology , Nails, Malformed/drug therapy , Nails, Malformed/pathology , Nails, Malformed/physiopathology , Osteochondrodysplasias/drug therapy , Osteochondrodysplasias/pathology , Osteochondrodysplasias/physiopathology , Potassium Channels/metabolism , Thumb/pathology , Thumb/physiopathology
14.
Exp Clin Endocrinol Diabetes ; 128(6-07): 414-422, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32242326

ABSTRACT

Despite its first description more than 75 years ago, effective treatment for "Allan-Herndon-Dudley-Syndrome (AHDS)", an X-linked thyroid hormone transporter defect, is unavailable. Mutations in the SLC16A2 gene have been discovered to be causative for AHDS in 2004, but a comprehensive understanding of the function of the encoded protein, monocarboxylate transporter 8 (MCT8), is incomplete. Patients with AHDS suffer from neurodevelopmental delay, as well as extrapyramidal (dystonia, chorea, athetosis), pyramidal (spasticity), and cerebellar symptoms (ataxia). This suggests an affection of the pyramidal tracts, basal ganglia, and cerebellum, most likely already during fetal brain development. The function of other brain areas relevant for mood, behavior, and vigilance seems to be intact. An optimal treatment strategy should thus aim to deliver T3 to these relevant structures at the correct time points during development. A potential therapeutic strategy meeting these needs might be the delivery of T3 via a "Trojan horse mechanism" by which T3 is delivered into target cells by a thyroid hormone transporter independent T3 internalization.


Subject(s)
Basal Ganglia , Cerebellum , Mental Retardation, X-Linked/drug therapy , Mental Retardation, X-Linked/metabolism , Mental Retardation, X-Linked/physiopathology , Monocarboxylic Acid Transporters/metabolism , Muscle Hypotonia/drug therapy , Muscle Hypotonia/metabolism , Muscle Hypotonia/physiopathology , Muscular Atrophy/drug therapy , Muscular Atrophy/metabolism , Muscular Atrophy/physiopathology , Pyramidal Tracts , Triiodothyronine/administration & dosage , Triiodothyronine/metabolism , Basal Ganglia/drug effects , Basal Ganglia/physiopathology , Cerebellum/drug effects , Cerebellum/physiopathology , Humans , Pyramidal Tracts/drug effects , Pyramidal Tracts/physiopathology
15.
Nat Commun ; 11(1): 480, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31980599

ABSTRACT

Mutations in the actively expressed, maternal allele of the imprinted KCNK9 gene cause Birk-Barel intellectual disability syndrome (BBIDS). Using a BBIDS mouse model, we identify here a partial rescue of the BBIDS-like behavioral and neuronal phenotypes mediated via residual expression from the paternal Kcnk9 (Kcnk9pat) allele. We further demonstrate that the second-generation HDAC inhibitor CI-994 induces enhanced expression from the paternally silenced Kcnk9 allele and leads to a full rescue of the behavioral phenotype suggesting CI-994 as a promising molecule for BBIDS therapy. Thus, these findings suggest a potential approach to improve cognitive dysfunction in a mouse model of an imprinting disorder.


Subject(s)
Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/metabolism , Histones/metabolism , Intellectual Disability/genetics , Intellectual Disability/metabolism , Muscle Hypotonia/genetics , Muscle Hypotonia/metabolism , Potassium Channels/genetics , Animals , Behavior, Animal , Benzamides , Brain/metabolism , Craniofacial Abnormalities/drug therapy , Disease Models, Animal , Female , Gene Knockdown Techniques , Genomic Imprinting , Histone Deacetylase Inhibitors/pharmacology , Humans , Intellectual Disability/drug therapy , Locus Coeruleus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Hypotonia/drug therapy , Mutation , Phenotype , Phenylenediamines/pharmacology , Potassium Channels/deficiency , Potassium Channels/metabolism , Up-Regulation/drug effects
16.
Lancet Diabetes Endocrinol ; 7(9): 695-706, 2019 09.
Article in English | MEDLINE | ID: mdl-31377265

ABSTRACT

BACKGROUND: Deficiency of the thyroid hormone transporter monocarboxylate transporter 8 (MCT8) causes severe intellectual and motor disability and high serum tri-iodothyronine (T3) concentrations (Allan-Herndon-Dudley syndrome). This chronic thyrotoxicosis leads to progressive deterioration in bodyweight, tachycardia, and muscle wasting, predisposing affected individuals to substantial morbidity and mortality. Treatment that safely alleviates peripheral thyrotoxicosis and reverses cerebral hypothyroidism is not yet available. We aimed to investigate the effects of treatment with the T3 analogue Triac (3,3',5-tri-iodothyroacetic acid, or tiratricol), in patients with MCT8 deficiency. METHODS: In this investigator-initiated, multicentre, open-label, single-arm, phase 2, pragmatic trial, we investigated the effectiveness and safety of oral Triac in male paediatric and adult patients with MCT8 deficiency in eight countries in Europe and one site in South Africa. Triac was administered in a predefined escalating dose schedule-after the initial dose of once-daily 350 µg Triac, the daily dose was increased progressively in 350 µg increments, with the goal of attaining serum total T3 concentrations within the target range of 1·4-2·5 nmol/L. We assessed changes in several clinical and biochemical signs of hyperthyroidism between baseline and 12 months of treatment. The prespecified primary endpoint was the change in serum T3 concentrations from baseline to month 12. The co-primary endpoints were changes in concentrations of serum thyroid-stimulating hormone (TSH), free and total thyroxine (T4), and total reverse T3 from baseline to month 12. These analyses were done in patients who received at least one dose of Triac and had at least one post-baseline evaluation of serum throid function. This trial is registered with ClinicalTrials.gov, number NCT02060474. FINDINGS: Between Oct 15, 2014, and June 1, 2017, we screened 50 patients, all of whom were eligible. Of these patients, four (8%) patients decided not to participate because of travel commitments. 46 (92%) patients were therefore enrolled in the trial to receive Triac (median age 7·1 years [range 0·8-66·8]). 45 (98%) participants received Triac and had at least one follow-up measurement of thyroid function and thus were included in the analyses of the primary endpoints. Of these 45 patients, five did not complete the trial (two patients withdrew [travel burden, severe pre-existing comorbidity], one was lost to follow-up, one developed of Graves disease, and one died of sepsis). Patients required a mean dose of 38.3 µg/kg of bodyweight (range 6·4-84·3) to attain T3 concentrations within the target range. Serum T3 concentration decreased from 4·97 nmol/L (SD 1·55) at baseline to 1·82 nmol/L (0·69) at month 12 (mean decrease 3·15 nmol/L, 95% CI 2·68-3·62; p<0·0001), while serum TSH concentrations decreased from 2·91 mU/L (SD 1·68) to 1·02 mU/L (1·14; mean decrease 1·89 mU/L, 1·39-2·39; p<0·0001) and serum free T4 concentrations decreased from 9·5 pmol/L (SD 2·5) to 3·4 (1·6; mean decrease 6·1 pmol/L (5·4-6·8; p<0·0001). Additionally, serum total T4 concentrations decreased by 31·6 nmol/L (28·0-35·2; p<0·0001) and reverse T3 by 0·08 nmol/L (0·05-0·10; p<0·0001). Seven treatment-related adverse events (transiently increased perspiration or irritability) occurred in six (13%) patients. 26 serious adverse events that were considered unrelated to treatment occurred in 18 (39%) patients (mostly hospital admissions because of infections). One patient died from pulmonary sepsis leading to multi-organ failure, which was unrelated to Triac treatment. INTERPRETATION: Key features of peripheral thyrotoxicosis were alleviated in paediatric and adult patients with MCT8 deficiency who were treated with Triac. Triac seems a reasonable treatment strategy to ameliorate the consequences of untreated peripheral thyrotoxicosis in patients with MCT8 deficiency. FUNDING: Dutch Scientific Organization, Sherman Foundation, NeMO Foundation, Wellcome Trust, UK National Institute for Health Research Cambridge Biomedical Centre, Toulouse University Hospital, and Una Vita Rara ONLUS.


Subject(s)
Membrane Transport Proteins/administration & dosage , Mental Retardation, X-Linked/drug therapy , Muscle Hypotonia/drug therapy , Muscular Atrophy/drug therapy , Triiodothyronine/analogs & derivatives , Adolescent , Child , Child, Preschool , Europe , Follow-Up Studies , Guidelines as Topic , Humans , Infant , Male , Membrane Transport Proteins/pharmacology , Mental Retardation, X-Linked/physiopathology , Muscle Hypotonia/physiopathology , Muscular Atrophy/physiopathology , Patient Safety , South Africa , Triiodothyronine/administration & dosage , Triiodothyronine/pharmacology , Young Adult
17.
Arch Argent Pediatr ; 117(4): e382-e386, 2019 08 01.
Article in Spanish | MEDLINE | ID: mdl-31339280

ABSTRACT

Nemaline myopathy (NM) is a heterogeneous disorder defined by the presence of rod-shaped structures known as nemaline bodies or rods. The diagnosis is based on muscle weakness, combined with visualization of nemaline bodies on muscle biopsy. There is no curative treatment for nemaline myopathy. Therapeutic strategies for this condition are symptomatic and empirical. Herein, we present a newborn with severe respiratory failure and generalized muscle weakness, who was diagnosed as NM by muscle biopsy. The patient experienced remarkable decrease in sialorrhea and improvement of spontaneous movements after L-tyrosine treatment. This case is presented to emphasize the importance of muscle biopsy in the differential diagnosis of severe hypotonia during neonatal period and a possible benefit of L-tyrosine supplementation for decreasing sialorrhea and restoring muscle strength.


La miopatía nemalínica es un trastorno heterogéneo definido por la presencia de estructuras con forma de bastones, conocidas como cuerpos nemalínicos (o bastones de nemalina). El diagnóstico se funda en la debilidad muscular, además de la visualización de cuerpos nemalínicos en la biopsia muscular. La miopatía nemalínica no tiene cura. Las estrategias terapéuticas para este trastorno son sintomáticas y empíricas. En este artículo, presentamos el caso de una recién nacida con insuficiencia respiratoria grave y debilidad muscular generalizada, a la que se le diagnosticó miopatía nemalínica a través de la biopsia muscular. La paciente tuvo una notable disminución de la sialorrea y una mejora de los movimientos espontáneos después del tratamiento con L-tirosina. Este caso se presenta para destacar la importancia de la biopsia muscular en el diagnóstico diferencial de la hipotonía grave durante el período neonatal y el posible beneficio del aporte suplementario de L-tirosina para disminuir la sialorrea y restaurar la fuerza muscular.


Subject(s)
Myopathies, Nemaline/drug therapy , Tyrosine/therapeutic use , Female , Humans , Infant, Newborn , Muscle Hypotonia/drug therapy , Muscle Hypotonia/etiology , Myopathies, Nemaline/complications , Sialorrhea/drug therapy , Sialorrhea/etiology , Treatment Outcome
18.
Pediatr Nephrol ; 34(10): 1727-1728, 2019 10.
Article in English | MEDLINE | ID: mdl-30903373

ABSTRACT

Hypomagnesemia with secondary hypocalcemia is a rare autosomal recessive disorder which manifests in early infancy with generalized seizures, other symptoms of neuromuscular irritability, and growth disturbances. Homozygous mutations in the magnesium transporter gene, transient receptor potential melastatin 6 (TRPM6), cause the disease. Here, we present an 8-month-old Turkish boy with a novel mutation of TRPM6. The patient, son of first-degree cousins, was hospitalized because of recurrent seizures and mild hypotonia. He had seizures since the newborn period and he had been treated with phenobarbital but there was no favorable response to therapy. His past history also revealed hypocalcemia detected on the newborn period but serum magnesium levels were not studied at that time. During hospitalization, we detected hypocalcemia, hypomagnesemia, and normal parathormone levels. Abdominal ultrasound was normal. Magnesium excretion was slightly increased. Considering the consanguinity of the parents and clinical features of the patients, genetic testing of the TRPM6 gene was performed and a novel homozygous mutation was detected as c.3178A>T. He was started on magnesium and calcium supplementation and he is symptom-free for 1 year. We would like to call attention to the measurement of serum magnesium levels in children with hypocalcemic convulsions. Early and appropriate treatment with magnesium supplementation is crucial.


Subject(s)
Calcium/administration & dosage , Hypocalcemia/etiology , Magnesium/blood , Muscle Hypotonia/etiology , Seizures/etiology , Calcium/blood , Humans , Hypocalcemia/blood , Hypocalcemia/diagnosis , Hypocalcemia/drug therapy , Infant , Magnesium/administration & dosage , Male , Muscle Hypotonia/blood , Muscle Hypotonia/diagnosis , Muscle Hypotonia/drug therapy , Phenobarbital/therapeutic use , Seizures/blood , Seizures/diagnosis , Seizures/drug therapy , Treatment Outcome
20.
Pan Afr Med J ; 30: 152, 2018.
Article in English | MEDLINE | ID: mdl-30374398

ABSTRACT

A vitamin B12 deficiency in infants is rare, but may sometimes be seen in breastfed babies of strict vegetarian mothers. Vitamin B12, also known as cobalamin, is only found in meat and other animal products. Most babies have a sufficient supply as long as the mother was not deficient herself. Symptoms and signs of vitamin B12 deficiency appear between the ages of 2 to 12 months and include vomiting, lethargy, failure to thrive, hypotonia, and arrest or regression of developmental skills. Urinary concentrations of methylmalonic acid and homocystine are characteristically elevated in vitamin B12 deficiency. Early treatment for a vitamin B12 deficiency in an infant involves immediate administration of vitamin B12 to the baby and the breastfeeding mother. The infant and mother will each receive an injection of vitamin B12 containing 1,000 mcg or more of the vitamin, and the mother will continue to receive injections every month to raise her own stores. After the initial injection, the baby will often receive future vitamin B12 through food sources. We present a case of vitamin B12 deficiency in a 9-month-old girl presented with psychomotor regression, hypotonia and lethargy. The child was exclusively breast-fed from birth by a mother who was on strict vegetarian diet and belong to a low socio-economic status. Laboratory data revealed bicytopenia with macrocytic anemia and methylmalonic acid in the urine, consistent with vitamin B12 deficient anemia. The Brain CT revealed a cerebral atrophy and delayed myelination. Vitamin B12 supply was effective on anaemia and psychomotor delay. This case figures out the importance of an early diagnosis in front of psychomoteur regression and hypotonia, given the risk of incomplete neurologic recovery due to vitamin B12 deficiency mainly in the setting of maternal nutritional deficiency.


Subject(s)
Anemia, Macrocytic/etiology , Psychomotor Disorders/etiology , Vitamin B 12 Deficiency/complications , Vitamin B 12/administration & dosage , Anemia, Macrocytic/diagnosis , Anemia, Macrocytic/drug therapy , Breast Feeding , Female , Humans , Infant , Mothers , Muscle Hypotonia/diagnosis , Muscle Hypotonia/drug therapy , Muscle Hypotonia/etiology , Psychomotor Disorders/drug therapy , Treatment Outcome , Vitamin B 12 Deficiency/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL
...