Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 444
Filter
1.
Cardiovasc Pathol ; 70: 107630, 2024.
Article in English | MEDLINE | ID: mdl-38490313

ABSTRACT

A female neonate born with normal Apgar scores at 38+2 weeks of gestational age unexpectedly passed away within less than 30 hours after birth. The situation mirrored her brother's earlier demise within 24 hours post-delivery, suggesting a possible genetic disorder. Gross examination revealed widespread cyanosis and distinct yellowish changes on the cardiac ventricles. Histopathological examination disclosed lipid accumulation in the liver, heart, and kidneys. Tandem mass spectrometry detected elevated levels of 10 amino acids and 14 carnitines in cardiac blood. Trio-whole genome sequencing (Trio-WGS) identified the SLC25A20 c.199-10T>G mutation associated with carnitine-acylcarnitine translocase disease (CACTD), a type of fatty acid oxidation disorders (FAODs) with a potential for sudden death. Further validation of gene expression confirmed the functional deficiency of SLC25A20, ultimately diagnosing CACTD as the underlying cause of the neonate's demise. This case highlights the importance of prenatal metabolic and genetic screening for prospective parents and emphasizes the need for forensic doctors to integrate metabolomic and genomic investigations into autopsies for suspected inherited metabolic diseases.


Subject(s)
Carnitine Acyltransferases , Lipid Metabolism, Inborn Errors , Mutation , Humans , Infant, Newborn , Female , Carnitine Acyltransferases/deficiency , Carnitine Acyltransferases/genetics , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Lipid Metabolism, Inborn Errors/complications , Lipid Metabolism, Inborn Errors/diagnosis , Phenotype , Fatal Outcome , Genetic Predisposition to Disease , Sudden Infant Death/genetics , Sudden Infant Death/pathology , Sudden Infant Death/etiology , Autopsy , Death, Sudden, Cardiac/etiology , Death, Sudden, Cardiac/pathology , Cause of Death , Carnitine/analogs & derivatives , Carnitine/deficiency , Mitochondrial Membrane Transport Proteins/genetics , Myocardium/pathology , Myocardium/metabolism , Membrane Transport Proteins
2.
Genet Med ; 26(6): 101123, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38501492

ABSTRACT

PURPOSE: Long-chain 3-hydroxyacyl-coenzyme A dehydrogenase deficiency (LCHADD) is a rare fatty acid oxidation disorder characterized by recurrent episodes of metabolic decompensation and rhabdomyolysis, as well as retinopathy, peripheral neuropathy, and cardiac involvement, such as infantile dilated cardiomyopathy. Because LCHADD patients are surviving longer, we sought to characterize LCHADD-associated major cardiac involvement in adolescence and young adulthood. METHODS: A retrospective cohort of 16 adolescent and young adult participants with LCHADD was reviewed for cardiac phenotype. RESULTS: Major cardiac involvement occurred in 9 of 16 participants, including sudden death, out-of-hospital cardiac arrest, acute cardiac decompensations with heart failure and/or in-hospital cardiac arrest, end-stage dilated cardiomyopathy, and moderate restrictive cardiomyopathy. Sudden cardiac arrest was more common in males and those with a history of infant cardiomyopathy. CONCLUSION: The cardiac manifestations of LCHADD in adolescence and early adulthood are complex and distinct from the phenotype seen in infancy. Life-threatening arrhythmia occurs at substantial rates in LCHADD, often in the absence of metabolic decompensation or rhabdomyolysis. The potential risk factors identified here-male sex and history of infant cardiomyopathy-may hint at strategies for risk stratification and possibly the prevention of these events.


Subject(s)
Lipid Metabolism, Inborn Errors , Phenotype , Humans , Male , Adolescent , Female , Young Adult , Adult , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Retrospective Studies , Rhabdomyolysis/genetics , Rhabdomyolysis/pathology , Rhabdomyolysis/enzymology , Long-Chain-3-Hydroxyacyl-CoA Dehydrogenase/genetics , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Death, Sudden, Cardiac/etiology , Death, Sudden, Cardiac/pathology
3.
Mol Genet Metab ; 142(1): 108351, 2024 May.
Article in English | MEDLINE | ID: mdl-38430613

ABSTRACT

Fatty acid oxidation disorders (FAODs) are a family of rare, genetic disorders that affect any part of the fatty acid oxidation pathway. Patients present with severe phenotypes, such as hypoketotic hypoglycemia, cardiomyopathy, and rhabdomyolysis, and currently manage these symptoms by the avoidance of fasting and maintaining a low-fat, high-carbohydrate diet. Because knowledge about FAODs is limited due to the small number of patients, rodent models have been crucial in learning more about these disorders, particularly in studying the molecular mechanisms involved in different phenotypes and in evaluating treatments for patients. The purpose of this review is to present the different FAOD mouse models and highlight the benefits and limitations of using these models. Specifically, we discuss the phenotypes of the available FAOD mouse models, the potential molecular causes of prominent FAOD phenotypes that have been studied using FAOD mouse models, and how FAOD mouse models have been used to evaluate treatments for patients.


Subject(s)
Disease Models, Animal , Fatty Acids , Lipid Metabolism, Inborn Errors , Oxidation-Reduction , Animals , Mice , Fatty Acids/metabolism , Humans , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/metabolism , Lipid Metabolism, Inborn Errors/pathology , Phenotype , Cardiomyopathies/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cardiomyopathies/etiology
4.
Neuromuscul Disord ; 35: 19-24, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38194732

ABSTRACT

The rare disorder known as Neutral Lipid Storage Disease with Myopathy presents with a variety of clinical manifestations, including myopathy, cardiac dysfunction, and other organ complications. Early diagnosis is crucial due to the increased risk of cardiomyopathy. We describe the clinical, histopathological, muscle imaging, and genetic findings of nine neutral lipid storage myopathy patients. Proximal weakness and asymmetric involvement may suggest lipid storage myopathy. While skeletal muscle weakness was the main manifestation in our patients, one case presented only with hyperCKemia. Additionally, three patients had fertility issues, two suffered from diabetes mellitus, two had cardiomyopathy, and one had a history of hypothyroidism. Muscle histopathology revealed lipid depositions and rimmed vacuoles, prompting peripheral blood smears to detect Jordan Anomalies. All muscle biopsies and peripheral blood smear showed lipid droplets, rimmed vacuoles, and Jordan anomaly. Identifying PNPLA2 gene mutations is important for diagnosing neutral lipid storage myopathy; our cases showed some novel mutations. This study highlights the importance of early diagnosis and comprehensive evaluation in managing neutral lipid storage myopathy cases.


Subject(s)
Cardiomyopathies , Ichthyosiform Erythroderma, Congenital , Lipid Metabolism, Inborn Errors , Muscular Diseases , Humans , Iran , Muscle, Skeletal/pathology , Lipase/genetics , Muscular Diseases/diagnosis , Muscular Diseases/genetics , Muscular Diseases/pathology , Lipid Metabolism, Inborn Errors/diagnosis , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Cardiomyopathies/diagnosis , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Mutation
5.
Microvasc Res ; 148: 104545, 2023 07.
Article in English | MEDLINE | ID: mdl-37146675

ABSTRACT

BACKGROUND: nailfold capillaroscopy (NCF) is a non-invasive imaging technique to seek peripheral microcirculation abnormalities in children and adults. Familial hypercholesterolemia is a genetic disorder caused by mutations capable of increasing blood levels of low-density lipoproteins cholesterol (LDL-C), thus triggering early atherosclerosis. The study aims at evaluating peripheral microcirculation in children with heterozygous familial hypercholesterolemia (HeFH) by means of NFC in comparison with healthy peers and at searching for possible correlations between these abnormalities and patients' lipid panel. METHODS: thirty-six HeFH patients were enrolled (13 males and 23 females. Mean age 8 ± 3 years; age range 3-13 years). They had increased levels of total cholesterol (237.9 ± 34.2 mg/dl) and LDL-C (154.2 ± 37.6 mg/dl). Both values were ≥95th gender and age specific centile. All the subjects in the study underwent NFC. RESULTS: In 69.4 % of HeFH children nailfold capillaries were tortuous (p < 0.00001 compared to healthy controls). In 41.6 % the number of capillaries was markedly reduced (<7 capillaries/mm). The mean number of capillaries was 8.4 ± 2.6/mm in HeFH and 12.2 ± 1.4/mm in healthy controls (p < 0.00001). In 100 % of the sample size capillary blood flow was slowed down (p < 0.00001). In 50 % of the sample size a blood "sludge" phenomenon was seen (p < 0.00001). No gender differences were detected. Sludge phenomenon was seen only in those with LDL-C over 99th centile (p < 0.00001). CONCLUSION: NCF allows the identification of an early peripheral microvascular dysfunction in HeFH children which is similar to that already seen in atherosclerotic disease. Prompt identification of these capillary abnormalities may be crucial in implementing early prevention measures.


Subject(s)
Capillaries , Lipid Metabolism, Inborn Errors , Microscopic Angioscopy , Humans , Male , Female , Child, Preschool , Child , Adolescent , Capillaries/diagnostic imaging , Capillaries/pathology , Microscopic Angioscopy/methods , Microcirculation , Lipid Metabolism, Inborn Errors/diagnostic imaging , Lipid Metabolism, Inborn Errors/pathology
6.
Pediatr Dermatol ; 40(5): 879-881, 2023.
Article in English | MEDLINE | ID: mdl-36709747

ABSTRACT

Chanarin-Dorfman syndrome (CDS) is a rare, autosomal recessive disorder of impaired triacylglycerol catabolism leading to cytoplasmic deposition of triglycerides in various cell types. We describe the case of an 8-month-old boy with cataracts, strabismus, motor delays, and an ichthyosiform rash since birth. Genetic testing revealed a pathogenic variant of the ABHD5 gene, suggestive of CDS, and further workup demonstrated hepatic steatosis and myopathy. His ichthyosis improved with initiation of a diet low in very long-chain fatty acids and medium-chain fatty acid supplementation.


Subject(s)
Cataract , Ichthyosiform Erythroderma, Congenital , Ichthyosis, Lamellar , Ichthyosis , Lipid Metabolism, Inborn Errors , Muscular Diseases , Male , Humans , Infant , Ichthyosiform Erythroderma, Congenital/diagnosis , Ichthyosiform Erythroderma, Congenital/genetics , Ichthyosis, Lamellar/diagnosis , Ichthyosis, Lamellar/genetics , Ichthyosis/diagnosis , Ichthyosis/genetics , Muscular Diseases/diagnosis , Muscular Diseases/genetics , Muscular Diseases/pathology , Lipid Metabolism, Inborn Errors/diagnosis , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Cataract/diagnosis , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics
7.
Clin Chim Acta ; 530: 39-44, 2022 May 01.
Article in English | MEDLINE | ID: mdl-35248527

ABSTRACT

BACKGROUND AND AIM: Clinical manifestations and genetic backgrounds of Japanese patients with sitosterolemia have been unclear. MATERIALS AND METHODS: We searched PubMed for studies using the keywords "sitosterolemia" or "phytosterolemia" and "Japan". Moreover, we added information from the members of the Committee on Primary Dyslipidemia under the Research Program on Rare and Intractable Disease of the Ministry of Health, Labour and Welfare (MHLW) of Japan. RESULTS: We identified 36 patients with sitosterolemia caused by biallelic pathogenic mutations in the ATP-binding cassette subfamily G member 5 (ABCG5) or ATP-binding cassette subfamily G member 8 (ABCG8) from 31 families in Japan. The diagnosed age ranged from 0 to 64 years (median 13 years). The median sitosterol and LDL cholesterol levels were 100 µg/ml (IQR: 50-183), and 193 mg/dl (IQR: 108-295), respectively. All the patients exhibited cutaneous and/or tendon xanthomas, up to 9 (25%) patients exhibited premature coronary artery disease, 5 (16%) patients exhibited arthritis, and 8 (22%) patients exhibited blood abnormalities. Ezetimibe was administered to all the patients, including infantile cases, while statins, colestimide, evolocumab, probucol, and LDL apheresis were also used. CONCLUSION: We are providing a demographic overview of the clinical and genetic backgrounds of Japanese patients with sitosterolemia.


Subject(s)
Intestinal Diseases , Lipid Metabolism, Inborn Errors , Phytosterols , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , Adenosine Triphosphate , Adolescent , Adult , Child , Child, Preschool , Humans , Hypercholesterolemia , Infant , Infant, Newborn , Intestinal Diseases/diagnosis , Intestinal Diseases/genetics , Intestinal Diseases/pathology , Japan , Lipid Metabolism, Inborn Errors/diagnosis , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Middle Aged , Phytosterols/adverse effects , Phytosterols/genetics , Young Adult
8.
Lipids Health Dis ; 21(1): 11, 2022 Jan 18.
Article in English | MEDLINE | ID: mdl-35042526

ABSTRACT

BACKGROUND: Sitosterolemia is a lipid disorder characterized by the accumulation of phytosterols in plasma and organs, caused by mutations in the ABCG5 and/or ABCG8 genes. The disease is frequently misdiagnosed and mistreated as familial hypercholesterolemia (FH). To gain a better understanding of the disease, the current status of diagnosis and treatment of Chinese patients with sitosterolemia was reviewed and summarized. METHOD: Literature search was performed. The clinical features and molecular characteristics of Chinese patients with sitosterolemia were analysed. Four children with sitosterolemia and the treatment experience were described. RESULTS: Fifty-five patients with sitosterolemia have been reported in China. These patients were aged from 3 months to 67 years at diagnosis, and the median was 8 years of age. Several complications, such as xanthomas in 47 patients (85%), thrombocytopenia in 17 patients (31%), anemia in 14 patients (25%), and cardiovascular damage in 12 patients (22%), were observed. Thirty-nine patients (71%) exhibited mutations in the ABCG5 gene, 15 patients (27%) showed mutations in ABCG8, and variations in both genes occurred in one patient (2%). A patient with two clinically rare diseases, namely, sitosterolemia and glycogen storage disease type VI (GSD VI)), is reported here for the first time. The four reported patients were treated with low cholesterol and phytosterol-limited diet alone or combined with cholestyramine. Even though decreases were observed for total plasma cholesterol (TC) and low-density-lipoprotein cholesterol (LDL-C), and these levels were as low as normal in some patients, the levels of plant sterols remained above the normal range. However, TC, LDL-C and plant sterol levels remained at high levels in patients treated with a control diet control only. CONCLUSIONS: The analysis reveals that different from Caucasians carrying mainly variations in ABCG8, most Chinese patients have mutations in the ABCG5 gene, and Arg446Ter, Gln251Ter, anArg389His might be hot-spot mutations in Chinese patients. The current survey provides clinical data to enable the development of a standardized protocol for the diagnosis and treatment of sitosterolemia in China.


Subject(s)
Hypercholesterolemia/diagnosis , Intestinal Diseases/diagnosis , Lipid Metabolism, Inborn Errors/diagnosis , Phytosterols/adverse effects , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , China , Female , Humans , Hypercholesterolemia/complications , Hypercholesterolemia/genetics , Hypercholesterolemia/pathology , Infant , Intestinal Diseases/complications , Intestinal Diseases/genetics , Intestinal Diseases/pathology , Lipid Metabolism, Inborn Errors/complications , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Lipoproteins/genetics , Male , Middle Aged , Mutation/genetics , Phytosterols/genetics , Young Adult
9.
Neuropathol Appl Neurobiol ; 48(1): e12731, 2022 02.
Article in English | MEDLINE | ID: mdl-33969514

ABSTRACT

Three consecutive skeletal muscle biopsies during a several months time-frame, showing different degrees of neutral lipid storage. This is highlighted by Oil-red-O stains (D, E, F) and electron microscopy (G, H, I). Note the impact on mitochondrial morphology with so called 'parking lots (K, L). Zooming 'in and out' into the ultrastructure, using the nanotomy platform provides interesting detailled information (http://nanotomy.org). ​.


Subject(s)
Lipid Metabolism, Inborn Errors , Muscular Diseases , Muscular Dystrophies , Humans , Immunoglobulins , Lipid Metabolism, Inborn Errors/pathology , Muscle, Skeletal/pathology , Muscular Diseases/pathology , Muscular Diseases/therapy , Plasmapheresis
10.
G3 (Bethesda) ; 12(2)2022 02 04.
Article in English | MEDLINE | ID: mdl-34791225

ABSTRACT

Ichthyoses are hereditary skin disorders characterized by the formation of scales and defects in the outermost layer of the epidermis. In dogs, at least six different breed-specific ichthyoses including a relatively common PNPLA1-related autosomal recessive ichthyosis in Golden Retrievers are known. In this study, we investigated 14 Golden Retrievers with scales that were not homozygous for the mutant PNPLA1 allele suggesting a genetically distinct new form of ichthyosis. Histopathological examinations showed lamellar, orthokeratotic hyperkeratosis, and mildly hyperplastic epidermis that led to the diagnosis of a nonepidermolytic ichthyosis. Combined linkage and homozygosity mapping in 14 cases and 30 nonaffected family members delimited a critical interval of ∼12.7 Mb on chromosome 23. Whole-genome sequencing of an affected dog revealed a single protein-changing variant within this region that was not present in 795 control genomes. The identified variant is a 14 bp deletion in the ABHD5 gene (c.1006_1019del), leading to a frameshift and altering the last 14 codons p.(Asp336Serfs*6). The genotypes at this variant showed perfect cosegregation with the ichthyosis phenotype in a large family comprising 14 cases and 72 controls. ABHD5 encodes an acyltransferase required for lipid metabolism. In humans, variants in ABHD5 cause Chanarin-Dorfman syndrome, a neutral lipid storage disease with ichthyosis. Our data in dogs together with the knowledge on the effects of ABHD5 variants in humans strongly suggest ABHD5:c.1006_1019del as candidate causative genetic variant for a new canine form of ichthyosis, which we propose to designate as Golden Retriever ichthyosis type 2 (ICH2).


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase , Ichthyosiform Erythroderma, Congenital , Ichthyosis, Lamellar , Ichthyosis , Lipid Metabolism, Inborn Errors , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , Animals , Dogs , Frameshift Mutation , Gene Deletion , Ichthyosiform Erythroderma, Congenital/genetics , Ichthyosiform Erythroderma, Congenital/pathology , Ichthyosis/genetics , Ichthyosis/pathology , Ichthyosis/veterinary , Ichthyosis, Lamellar/genetics , Ichthyosis, Lamellar/veterinary , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Plant Breeding
11.
Int J Mol Sci ; 22(19)2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34638902

ABSTRACT

Medium-chain fatty acids (mc-FAs) are currently applied in the treatment of long-chain fatty acid oxidation disorders (lc-FAOD) characterized by impaired ß-oxidation. Here, we performed lipidomic and proteomic analysis in fibroblasts from patients with very long-chain acyl-CoA dehydrogenase (VLCADD) and long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHADD) deficiencies after incubation with heptanoate (C7) and octanoate (C8). Defects of ß-oxidation induced striking proteomic alterations, whereas the effect of treatment with mc-FAs was minor. However, mc-FAs induced a remodeling of complex lipids. Especially C7 appeared to act protectively by restoring sphingolipid biosynthesis flux and improving the observed dysregulation of protein homeostasis in LCHADD under control conditions.


Subject(s)
Caprylates/pharmacology , Fibroblasts/drug effects , Heptanoates/pharmacology , Lipid Metabolism, Inborn Errors/metabolism , Lipidomics/methods , Proteomics/methods , Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Cardiolipins/metabolism , Cell Line , Female , Fibroblasts/metabolism , Genotype , Humans , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Male , Mitochondria/drug effects , Mitochondria/metabolism , Phosphatidylcholines/metabolism , Phosphatidylethanolamines/metabolism , Proteome/metabolism , Sphingolipids/metabolism
12.
Cell Mol Life Sci ; 78(14): 5631-5646, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34110423

ABSTRACT

Peroxisomes play an essential role in the ß-oxidation of dicarboxylic acids (DCAs), which are metabolites formed upon ω-oxidation of fatty acids. Genetic evidence linking transporters and enzymes to specific DCA ß-oxidation steps is generally lacking. Moreover, the physiological functions of DCA metabolism remain largely unknown. In this study, we aimed to characterize the DCA ß-oxidation pathway in human cells, and to evaluate the biological role of DCA metabolism using mice deficient in the peroxisomal L-bifunctional protein (Ehhadh KO mice). In vitro experiments using HEK-293 KO cell lines demonstrate that ABCD3 and ACOX1 are essential in DCA ß-oxidation, whereas both the bifunctional proteins (EHHADH and HSD17B4) and the thiolases (ACAA1 and SCPx) have overlapping functions and their contribution may depend on expression level. We also show that medium-chain 3-hydroxydicarboxylic aciduria is a prominent feature of EHHADH deficiency in mice most notably upon inhibition of mitochondrial fatty acid oxidation. Using stable isotope tracing methodology, we confirmed that products of peroxisomal DCA ß-oxidation can be transported to mitochondria for further metabolism. Finally, we show that, in liver, Ehhadh KO mice have increased mRNA and protein expression of cholesterol biosynthesis enzymes with decreased (in females) or similar (in males) rate of cholesterol synthesis. We conclude that EHHADH plays an essential role in the metabolism of medium-chain DCAs and postulate that peroxisomal DCA ß-oxidation is a regulator of hepatic cholesterol biosynthesis.


Subject(s)
Cholesterol/metabolism , Dicarboxylic Acids/urine , Lipid Metabolism, Inborn Errors/pathology , Liver Diseases/pathology , Mitochondria/pathology , Peroxisomal Bifunctional Enzyme/physiology , Animals , Female , HEK293 Cells , Homeostasis , Humans , Lipid Metabolism, Inborn Errors/etiology , Liver Diseases/etiology , Liver Diseases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism
13.
Int J Mol Sci ; 22(5)2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33807969

ABSTRACT

Sitosterolemia is a lipid disorder characterized by the accumulation of dietary xenosterols in plasma and tissues caused by mutations in either ABCG5 or ABCG8. ABCG5 ABCG8 encodes a pair of ABC half transporters that form a heterodimer (G5G8), which then traffics to the surface of hepatocytes and enterocytes and promotes the secretion of cholesterol and xenosterols into the bile and the intestinal lumen. We review the literature from the initial description of the disease, the discovery of its genetic basis, current therapy, and what has been learned from animal, cellular, and molecular investigations of the transporter in the twenty years since its discovery. The genomic era has revealed that there are far more carriers of loss of function mutations and likely pathogenic variants of ABCG5 ABCG8 than previously thought. The impact of these variants on G5G8 structure and activity are largely unknown. We propose a classification system for ABCG5 ABCG8 mutants based on previously published systems for diseases caused by defects in ABC transporters. This system establishes a framework for the comprehensive analysis of disease-associated variants and their impact on G5G8 structure-function.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , Cholesterol/metabolism , Hypercholesterolemia , Intestinal Diseases , Lipid Metabolism, Inborn Errors , Lipoproteins , Mutation , Phytosterols/adverse effects , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 5/history , ATP Binding Cassette Transporter, Subfamily G, Member 5/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 8/history , ATP Binding Cassette Transporter, Subfamily G, Member 8/metabolism , Animals , Enterocytes/metabolism , Enterocytes/pathology , Hepatocytes/metabolism , Hepatocytes/pathology , History, 21st Century , Humans , Hypercholesterolemia/genetics , Hypercholesterolemia/history , Hypercholesterolemia/metabolism , Hypercholesterolemia/pathology , Intestinal Diseases/genetics , Intestinal Diseases/history , Intestinal Diseases/metabolism , Intestinal Diseases/pathology , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/history , Lipid Metabolism, Inborn Errors/metabolism , Lipid Metabolism, Inborn Errors/pathology , Lipoproteins/genetics , Lipoproteins/history , Lipoproteins/metabolism , Phytosterols/genetics , Phytosterols/history , Phytosterols/metabolism
15.
Int J Mol Sci ; 22(7)2021 Apr 06.
Article in English | MEDLINE | ID: mdl-33917608

ABSTRACT

In general, metabolic flexibility refers to an organism's capacity to adapt to metabolic changes due to differing energy demands. The aim of this work is to summarize and discuss recent findings regarding variables that modulate energy regulation in two different pathways of mitochondrial fatty metabolism: ß-oxidation and fatty acid biosynthesis. We focus specifically on two diseases: very long-chain acyl-CoA dehydrogenase deficiency (VLCADD) and malonyl-CoA synthetase deficiency (acyl-CoA synthetase family member 3 (ACSF3)) deficiency, which are both characterized by alterations in metabolic flexibility. On the one hand, in a mouse model of VLCAD-deficient (VLCAD-/-) mice, the white skeletal muscle undergoes metabolic and morphologic transdifferentiation towards glycolytic muscle fiber types via the up-regulation of mitochondrial fatty acid biosynthesis (mtFAS). On the other hand, in ACSF3-deficient patients, fibroblasts show impaired mitochondrial respiration, reduced lipoylation, and reduced glycolytic flux, which are compensated for by an increased ß-oxidation rate and the use of anaplerotic amino acids to address the energy needs. Here, we discuss a possible co-regulation by mtFAS and ß-oxidation in the maintenance of energy homeostasis.


Subject(s)
Congenital Bone Marrow Failure Syndromes/metabolism , Fatty Acids/metabolism , Lipid Metabolism, Inborn Errors/metabolism , Lipogenesis , Metabolic Diseases/metabolism , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Muscular Diseases/metabolism , Animals , Bacterial Proteins/metabolism , Coenzyme A Ligases/deficiency , Coenzyme A Ligases/metabolism , Congenital Bone Marrow Failure Syndromes/genetics , Congenital Bone Marrow Failure Syndromes/pathology , Fatty Acids/genetics , Humans , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Metabolic Diseases/genetics , Metabolic Diseases/pathology , Mice , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Muscular Diseases/genetics , Muscular Diseases/pathology
16.
Mol Genet Metab ; 133(1): 1-7, 2021 05.
Article in English | MEDLINE | ID: mdl-33744096

ABSTRACT

Mutations in the HADHB gene lead to Mitochondrial Trifunctional Protein (MTP) deficiency. MTP deficiency is a rare autosomal recessive disorder affecting long-chain fatty acid oxidation. Patients affected by MTP deficiency are unable to metabolize long-chain fatty-acids and suffer a variety of symptoms exacerbated during fasting. The three phenotypes associated with complete MTP deficiency are an early-onset cardiomyopathy and early death, an intermediate form with recurrent hypoketotic hypoglycemia and a sensorimotor neuropathy with episodic rhabdomyolysis with small amount of residual enzyme activities. This review aims to discuss the pathophysiological mechanisms and clinical manifestations of each phenotype, which appears different and linked to HADHB expression levels. Notably, the pathophysiology of the sensorimotor neuropathy is relatively unknown and we provide a hypothesis on the qualitative aspect of the role of acylcarnitine buildup in Schwann cells in MTP deficiency patients. We propose that acylcarnitine may exit the Schwann cell and alter membrane properties of nearby axons leading to axonal degeneration based on recent findings in different metabolic disorders.


Subject(s)
Cardiomyopathies/genetics , Lipid Metabolism, Inborn Errors/genetics , Mitochondrial Myopathies/genetics , Mitochondrial Trifunctional Protein, beta Subunit/genetics , Mitochondrial Trifunctional Protein/deficiency , Mitochondrial Trifunctional Protein/genetics , Nervous System Diseases/genetics , Rhabdomyolysis/genetics , Cardiomyopathies/pathology , Humans , Lipid Metabolism, Inborn Errors/pathology , Mitochondrial Myopathies/pathology , Mutation/genetics , Nervous System Diseases/pathology , Phenotype , Rhabdomyolysis/pathology
17.
Genes (Basel) ; 12(2)2021 02 17.
Article in English | MEDLINE | ID: mdl-33671400

ABSTRACT

Defects in the mitochondrial genome (mitochondrial DNA (mtDNA)) are associated with both congenital and acquired disorders in humans. Nuclear-encoded DNA polymerase subunit gamma (POLG) plays an important role in mtDNA replication, and proofreading and mutations in POLG have been linked with increased mtDNA deletions. SSBP1 is also a crucial gene for mtDNA replication. Here, we describe a patient diagnosed with Pearson syndrome with large mtDNA deletions that were not detected in the somatic cells of the mother. Exome sequencing was used to evaluate the nuclear factors associated with the patient and his family, which revealed a paternal POLG mutation (c.868C > T) and a maternal SSBP1 mutation (c.320G > A). The patient showed lower POLG and SSBP1 expression than his healthy brothers and the general population of a similar age. Notably, c.868C in the wild-type allele was highly methylated in the patient compared to the same site in both his healthy brothers. These results suggest that the co- deficient expression of POLG and SSBP1 genes could contribute to the development of mtDNA deletion.


Subject(s)
Congenital Bone Marrow Failure Syndromes/genetics , DNA Polymerase gamma/genetics , DNA, Mitochondrial/genetics , DNA-Binding Proteins/genetics , Genetic Predisposition to Disease , Lipid Metabolism, Inborn Errors/genetics , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Muscular Diseases/genetics , Adolescent , Adult , Child , Child, Preschool , Congenital Bone Marrow Failure Syndromes/pathology , DNA Replication/genetics , Female , Humans , Lipid Metabolism, Inborn Errors/pathology , Male , Mitochondrial Diseases/pathology , Muscular Diseases/pathology , Pedigree , Sequence Deletion/genetics , Exome Sequencing
18.
J Inherit Metab Dis ; 44(4): 893-902, 2021 07.
Article in English | MEDLINE | ID: mdl-33638202

ABSTRACT

Peripheral neuropathy is a known irreversible long-term complication of long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD) and mitochondrial trifunctional protein deficiency (MTPD), two inherited disorders of mitochondrial long-chain fatty acid oxidation. The underlying pathophysiology of neuropathy is still not fully understood. We report electrophysiological studies and neurological findings in a series of 8 LCHAD-deficient and 11 MTP-deficient patients. The median age at time of the study was 8.0 years (0.5-25 years). The overall prevalence of neuropathy was 58% with neuropathic symptoms being slightly more common in MTPD compared to LCHADD (70% vs 50%, respectively). Onset of neuropathy was significantly earlier in MTPD patients compared to LCHADD patients (median age at onset 4.7 vs 15.3 years, respectively, P = .047). In four patients, isolated peripheral neuropathy was the first and only presenting symptom, and in all four the diagnosis was missed by newborn screening. About half of the patients (45.5%) had a sensorimotor neuropathy, while 27.3% showed a pure motor form and another 27.3% an isolated sensory form. Despite early diagnosis by newborn screening and early initiation of therapy, peripheral neuropathy cannot be prevented in all patients with LCHADD/MTPD and has severe impact on the life of affected patients. Electrophysiology classifies LCHADD/MTPD neuropathy as axonal with secondary demyelination. A novel observation is that in patients with acute, fulminant onset of neuropathy, symptoms can be partly reversible. Further studies are needed to elucidate the underlying pathophysiology of axonal damage and possible therapeutic targets.


Subject(s)
Cardiomyopathies/complications , Lipid Metabolism, Inborn Errors/complications , Mitochondrial Myopathies/complications , Mitochondrial Trifunctional Protein/deficiency , Nervous System Diseases/complications , Peripheral Nervous System Diseases/diagnosis , Peripheral Nervous System Diseases/etiology , Rhabdomyolysis/complications , Adolescent , Adult , Age Factors , Cardiomyopathies/diagnosis , Cardiomyopathies/pathology , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Lipid Metabolism, Inborn Errors/diagnosis , Lipid Metabolism, Inborn Errors/pathology , Male , Mitochondrial Myopathies/diagnosis , Mitochondrial Myopathies/pathology , Nervous System Diseases/diagnosis , Nervous System Diseases/pathology , Peripheral Nervous System Diseases/pathology , Phenotype , Rhabdomyolysis/diagnosis , Rhabdomyolysis/pathology , Young Adult
19.
Biochim Biophys Acta Mol Basis Dis ; 1867(6): 166100, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33549744

ABSTRACT

Very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency is the most common inborn long-chain fatty acid oxidation (FAO) disorder. VLCAD deficiency is characterized by distinct phenotypes. The severe phenotypes are potentially life-threatening and affect the heart or liver, with a comparatively milder phenotype characterized by myopathic symptoms. There is an unmet clinical need for effective treatment options for the myopathic phenotype. The molecular mechanisms driving the gradual decrease in mitochondrial function and associated alterations of muscle fibers are unclear. The peroxisome proliferator-activated receptor (PPAR) pan-agonist bezafibrate is a potent modulator of FAO and multiple other mitochondrial functions and has been proposed as a potential medication for myopathic cases of long-chain FAO disorders. In vitro experiments have demonstrated the ability of bezafibrate to increase VLCAD expression and activity. However, the outcome of small-scale clinical trials has been controversial. We found VLCAD deficient patient fibroblasts to have an increased oxidative stress burden and deranged mitochondrial bioenergetic capacity, compared to controls. Applying heat stress under fasting conditions to bezafibrate pretreated patient cells, caused a marked further increase of mitochondrial superoxide levels. Patient cells failed to maintain levels of the essential thiol peptide antioxidant glutathione and experienced a decrease in cellular viability. Our findings indicate that chronic PPAR activation is a plausible initiator of long-term pathogenesis in VLCAD deficiency. Our findings further implicate disruption of redox homeostasis as a key pathogenic mechanism in VLCAD deficiency and support the notion that a deranged thiol metabolism might be an important pathogenic factor in VLCAD deficiency.


Subject(s)
Bezafibrate/pharmacology , Congenital Bone Marrow Failure Syndromes/drug therapy , Energy Metabolism , Fibroblasts/drug effects , Hypolipidemic Agents/pharmacology , Lipid Metabolism, Inborn Errors/drug therapy , Mitochondria/drug effects , Mitochondrial Diseases/drug therapy , Muscular Diseases/drug therapy , Peroxisome Proliferator-Activated Receptors/metabolism , Congenital Bone Marrow Failure Syndromes/metabolism , Congenital Bone Marrow Failure Syndromes/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Lipid Metabolism, Inborn Errors/metabolism , Lipid Metabolism, Inborn Errors/pathology , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Muscular Diseases/metabolism , Muscular Diseases/pathology , Oxidative Stress , Peroxisome Proliferator-Activated Receptors/genetics
20.
Neuromuscul Disord ; 31(1): 52-55, 2021 01.
Article in English | MEDLINE | ID: mdl-33303358

ABSTRACT

Neutral lipid storage disease with myopathy is an ultra-rare, inherited autosomal recessive neuromuscular metabolic disorder caused by pathogenic variants in PNPLA2. It typically presents in adults as a progressive myopathy and is associated with myocardiopathy, hepatic involvement, and high creatine kinase levels. Only three children and adolescents with neutral lipid storage disease with myopathy have been reported. We report a female infant with congenital hypotonia born to consanguineous parents, whose mother presented with polyhydramnios during pregnancy. She demonstrated delayed acquisition of motor milestones, hepatomegaly, and elevated creatine kinase levels. Homozygous pathogenic variants in PNPLA2 were identified. Lipid accumulation was observed within the muscle fibers and Jordans' anomaly was observed in a blood smear. This is the first report to describe an infant with mildly symptomatic neutral lipid storage disease with myopathy and demonstrate hepatic involvement in a pediatric patient. Despite her mild symptoms, her ancillary test results were markedly abnormal.


Subject(s)
Hepatomegaly/etiology , Lipid Metabolism, Inborn Errors/diagnosis , Muscle Hypotonia/etiology , Muscular Diseases/diagnosis , Adult , Child, Preschool , Female , Humans , Infant , Lipid Metabolism, Inborn Errors/pathology , Male , Muscular Diseases/pathology , Mutation , Pedigree , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...