1
|
DeVine T, Elizondo G, Gaston G, Babcock SJ, Matern D, Shchepinov MS, Pennesi ME, Harding CO, Gillingham MB. iPSC-Derived LCHADD Retinal Pigment Epithelial Cells Are Susceptible to Lipid Peroxidation and Rescued by Transfection of a Wildtype AAV-HADHA Vector. Invest Ophthalmol Vis Sci 2024; 65:22. [PMID: 39283617 PMCID: PMC11407479 DOI: 10.1167/iovs.65.11.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
Purpose Progressive choroid and retinal pigment epithelial (RPE) degeneration causing vision loss is a unique characteristic of long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD), a fatty acid oxidation disorder caused by a common c.1528G>C pathogenic variant in HADHA, the α subunit of the mitochondrial trifunctional protein (TFP). We established and characterized an induced pluripotent stem cell (iPSC)-derived RPE cell model from cultured skin fibroblasts of patients with LCHADD and tested whether addition of wildtype (WT) HAHDA could rescue the phenotypes identified in LCHADD-RPE. Methods We constructed an rAAV expression vector containing 3' 3xFLAG-tagged human HADHA cDNA under the transcriptional control of the cytomegalovirus (CMV) enhancer-chicken beta actin (CAG) promoter (CAG-HADHA-3XFLAG). LCHADD-RPE were cultured, matured, and transduced with either AAV-GFP (control) or AAV-HADHA-3XFLAG. Results LCHADD-RPE express TFP subunits and accumulate 3-hydroxy-acylcarnitines, cannot oxidize palmitate, and release fewer ketones than WT-RPE. When LCHADD-RPE are exposed to docosahexaenoic acid (DHA), they have increased oxidative stress, lipid peroxidation, decreased viability, and are rescued by antioxidant agents potentially explaining the pathologic mechanism of RPE loss in LCHADD. Transduced LCHADD-RPE expressing a WT copy of TFPα incorporated TFPα-FLAG into the TFP complex in the mitochondria and accumulated significantly less 3-hydroxy-acylcarnitines, released more ketones in response to palmitate, and were more resistant to oxidative stress following DHA exposure than control. Conclusions iPSC-derived LCHADD-RPE are susceptible to lipid peroxidation mediated cell death and are rescued by exogenous HADHA delivered with rAAV. These results are promising for AAV-HADHA gene addition therapy as a possible treatment for chorioretinopathy in patients with LCHADD.
Collapse
Affiliation(s)
- Tiffany DeVine
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon, United States
| | - Gabriela Elizondo
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon, United States
| | - Garen Gaston
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon, United States
| | - Shannon J Babcock
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon, United States
| | - Dietrich Matern
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, United States
| | - Mikhail S Shchepinov
- Institut des Biomolécules Max Mousseron, Université de Montpellier, CNRS, ENSCM, Montpellier, France
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Mark E Pennesi
- Ophthalmic Genetics Service, Casey Eye Institute, Oregon Health and Science University, Portland, Oregon, United States
- Retina Foundation of the Southwest, Dallas, Texas, United States
| | - Cary O Harding
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon, United States
| | - Melanie B Gillingham
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon, United States
| |
Collapse
|
2
|
Maurya M, Liu CH, Bora K, Kushwah N, Pavlovich MC, Wang Z, Chen J. Animal Models of Retinopathy of Prematurity: Advances and Metabolic Regulators. Biomedicines 2024; 12:1937. [PMID: 39335451 PMCID: PMC11428941 DOI: 10.3390/biomedicines12091937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/08/2024] [Accepted: 08/15/2024] [Indexed: 09/30/2024] Open
Abstract
Retinopathy of prematurity (ROP) is a primary cause of visual impairment and blindness in premature newborns, characterized by vascular abnormalities in the developing retina, with microvascular alteration, neovascularization, and in the most severe cases retinal detachment. To elucidate the pathophysiology and develop therapeutics for ROP, several pre-clinical experimental models of ROP were developed in different species. Among them, the oxygen-induced retinopathy (OIR) mouse model has gained the most popularity and critically contributed to our current understanding of pathological retinal angiogenesis and the discovery of potential anti-angiogenic therapies. A deeper comprehension of molecular regulators of OIR such as hypoxia-inducible growth factors including vascular endothelial growth factors as primary perpetrators and other new metabolic modulators such as lipids and amino acids influencing pathological retinal angiogenesis is also emerging, indicating possible targets for treatment strategies. This review delves into the historical progressions that gave rise to the modern OIR models with a focus on the mouse model. It also reviews the fundamental principles of OIR, recent advances in its automated assessment, and a selected summary of metabolic investigation enabled by OIR models including amino acid transport and metabolism.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jing Chen
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
3
|
Peña-Quintana L, Correcher-Medina P. Nutritional Management of Patients with Fatty Acid Oxidation Disorders. Nutrients 2024; 16:2707. [PMID: 39203843 PMCID: PMC11356788 DOI: 10.3390/nu16162707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 09/03/2024] Open
Abstract
Treatment of fatty acid oxidation disorders is based on dietary, pharmacological and metabolic decompensation measures. It is essential to provide the patient with sufficient glucose to prevent lipolysis and to avoid the use of fatty acids as fuel as far as possible. Dietary management consists of preventing periods of fasting and restricting fat intake by increasing carbohydrate intake, while maintaining an adequate and uninterrupted caloric intake. In long-chain deficits, long-chain triglyceride restriction should be 10% of total energy, with linoleic acid and linolenic acid intake of 3-4% and 0.5-1% (5/1-10/1 ratio), with medium-chain triglyceride supplementation at 10-25% of total energy (total MCT+LCT ratio = 20-35%). Trihepatnoin is a new therapeutic option with a good safety and efficacy profile. Patients at risk of rhabdomyolysis should ingest MCT or carbohydrates or a combination of both 20 min before exercise. In medium- and short-chain deficits, dietary modifications are not advised (except during exacerbations), with MCT contraindicated and slow sugars recommended 20 min before any significant physical exertion. Parents should be alerted to the need to increase the amount and frequency of carbohydrate intake in stressful situations. The main measure in emergency hospital treatment is the administration of IV glucose. The use of carnitine remains controversial and new therapeutic options are under investigation.
Collapse
Affiliation(s)
- Luis Peña-Quintana
- Pediatric Gastroenterology and Nutrition Unit, Insular Materno-Infantil University Hospital Complex, Asociación Canaria de Investigación Pediátrica, Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición ISCIII, University of Las Palmas de Gran Canaria, 35016 Las Palmas de Gran Canaria, Spain
- Asociación Española para el Estudio de los Errores Congénitos del Metabolismo (AECOM), 28221 Majadahonda, Spain
| | - Patricia Correcher-Medina
- Asociación Española para el Estudio de los Errores Congénitos del Metabolismo (AECOM), 28221 Majadahonda, Spain
- Metabolic and Nutrition Unit, Hospital Universitari i Politècnic la Fe, 46026 Valencia, Spain
| |
Collapse
|
4
|
Liepinsh E, Zvejniece L, Clemensson L, Ozola M, Vavers E, Cirule H, Korzh S, Skuja S, Groma V, Briviba M, Grinberga S, Liu W, Olszewski P, Gentreau M, Fredriksson R, Dambrova M, Schiöth HB. Hydroxymethylglutaryl-CoA reductase activity is essential for mitochondrial β-oxidation of fatty acids to prevent lethal accumulation of long-chain acylcarnitines in the mouse liver. Br J Pharmacol 2024; 181:2750-2773. [PMID: 38641905 DOI: 10.1111/bph.16363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 01/16/2024] [Accepted: 01/30/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND AND PURPOSE Statins are competitive inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase (HMGCR), and exert adverse effects on mitochondrial function, although the mechanisms underlying these effects remain unclear. We used a tamoxifen-induced Hmgcr-knockout (KO) mouse model, a multi-omics approach and mitochondrial function assessments to investigate whether decreased HMGCR activity impacts key liver energy metabolism pathways. EXPERIMENTAL APPROACH We established a new mouse strain using the Cre/loxP system, which enabled whole-body deletion of Hmgcr expression. These mice were crossed with Rosa26Cre mice and treated with tamoxifen to delete Hmgcr in all cells. We performed transcriptomic and metabolomic analyses and thus evaluated time-dependent changes in metabolic functions to identify the pathways leading to cell death in Hmgcr-KO mice. KEY RESULTS Lack of Hmgcr expression resulted in lethality, due to acute liver damage caused by rapid disruption of mitochondrial fatty acid β-oxidation and very high accumulation of long-chain (LC) acylcarnitines in both male and female mice. Gene expression and KO-related phenotype changes were not observed in other tissues. The progression to liver failure was driven by diminished peroxisome formation, which resulted in impaired mitochondrial and peroxisomal fatty acid metabolism, enhanced glucose utilization and whole-body hypoglycaemia. CONCLUSION AND IMPLICATIONS Our findings suggest that HMGCR is crucial for maintaining energy metabolism balance, and its activity is necessary for functional mitochondrial β-oxidation. Moreover, statin-induced adverse reactions might be rescued by the prevention of LC acylcarnitine accumulation.
Collapse
Affiliation(s)
- Edgars Liepinsh
- Latvian Institute of Organic Synthesis, Riga, Latvia
- Riga Stradins University, Riga, Latvia
| | | | | | - Melita Ozola
- Latvian Institute of Organic Synthesis, Riga, Latvia
- Riga Stradins University, Riga, Latvia
| | - Edijs Vavers
- Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Helena Cirule
- Latvian Institute of Organic Synthesis, Riga, Latvia
| | | | | | | | - Monta Briviba
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | | | - Wen Liu
- Uppsala University, Uppsala, Sweden
| | | | | | | | - Maija Dambrova
- Latvian Institute of Organic Synthesis, Riga, Latvia
- Riga Stradins University, Riga, Latvia
| | | |
Collapse
|
5
|
Gillingham MB, Choi D, Gregor A, Wongchaisuwat N, Black D, Scanga HL, Nischal KK, Sahel JA, Arnold G, Vockley J, Harding CO, Pennesi ME. Early diagnosis and treatment by newborn screening (NBS) or family history is associated with improved visual outcomes for long-chain 3-hydroxyacylCoA dehydrogenase deficiency (LCHADD) chorioretinopathy. J Inherit Metab Dis 2024; 47:746-756. [PMID: 38623632 PMCID: PMC11251862 DOI: 10.1002/jimd.12738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/17/2024]
Abstract
Long chain 3-hydroxyacyl-CoA dehydrogenase (LCHADD) is the only fatty acid oxidation disorder to develop a progressive chorioretinopathy resulting in vision loss; newborn screening (NBS) for this disorder began in the United States around 2004. We compared visual outcomes among 40 participants with LCHADD or trifunctional protein deficiency diagnosed symptomatically to those who were diagnosed via NBS or a family history. Participants completed ophthalmologic testing including measures of visual acuity, electroretinograms (ERG), fundal imaging, contrast sensitivity, and visual fields. Records were reviewed to document medical and treatment history. Twelve participants presented symptomatically with hypoglycemia, failure to thrive, liver dysfunction, cardiac arrest, or rhabdomyolysis. Twenty eight were diagnosed by NBS or due to a family history of LCHADD. Participants diagnosed symptomatically were older but had similar percent males and genotypes as those diagnosed by NBS. Treatment consisted of fasting avoidance, dietary long-chain fat restriction, MCT, C7, and/or carnitine supplementation. Visual acuity, rod- and cone-driven amplitudes on ERG, contrast sensitivity scores, and visual fields were all significantly worse among participants diagnosed symptomatically compared to NBS. In mixed-effects models, both age and presentation (symptomatic vs. NBS) were significant independent factors associated with visual outcomes. This suggests that visual outcomes were improved by NBS, but there was still lower visual function with advancing age in both groups. Early diagnosis and treatment by NBS is associated with improved visual outcomes and retinal function compared to participants who presented symptomatically. Despite the impact of early intervention, chorioretinopathy was greater with advancing age, highlighting the need for novel treatments.
Collapse
Affiliation(s)
- Melanie B Gillingham
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, USA
| | - Dongseok Choi
- OHSU-PSU School of Public Health, Biostatistics, Oregon Health & Science University, Portland, Oregon, USA
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Ashley Gregor
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, USA
| | - Nida Wongchaisuwat
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Danielle Black
- Division of Genetic and Genomic Medicine, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hannah L Scanga
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ken K Nischal
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jose-Alain Sahel
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Georgianne Arnold
- Division of Genetic and Genomic Medicine, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jerry Vockley
- Division of Genetic and Genomic Medicine, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Cary O Harding
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, USA
| | - Mark E Pennesi
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
6
|
Babcock SJ, Curtis AG, Gaston G, Elizondo G, Gillingham MB, Ryals RC. The LCHADD Mouse Model Recapitulates Early-Stage Chorioretinopathy in LCHADD Patients. Invest Ophthalmol Vis Sci 2024; 65:33. [PMID: 38904639 PMCID: PMC11193142 DOI: 10.1167/iovs.65.6.33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/27/2024] [Indexed: 06/22/2024] Open
Abstract
Purpose Recent studies have shown that the retinal pigment epithelium (RPE) relies on fatty acid oxidation (FAO) for energy, however, its role in overall retinal health is unknown. The only FAO disorder that presents with chorioretinopathy is long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD). Studying the molecular mechanisms can lead to new treatments for patients and elucidate the role of FAO in the RPE. This paper characterizes the chorioretinopathy progression in a recently reported LCHADD mouse model. Methods Visual assessments, such as optokinetic tracking and fundus imaging, were performed in wildtype (WT) and LCHADD mice at 3, 6, 10, and 12 months of age. Retinal morphology was analyzed in 12-month retinal cross-sections using hematoxylin and eosin (H&E), RPE65, CD68, and TUNEL staining, whereas RPE structure was assessed using transmission electron microscopy (TEM). Acylcarnitine profiles were measured in isolated RPE/sclera samples to determine if FAO was blocked. Bulk RNA-sequencing of 12 month old male WT mice and LCHADD RPE/sclera samples assessed gene expression changes. Results LCHADD RPE/sclera samples had a 5- to 7-fold increase in long-chain hydroxyacylcarnitines compared to WT, suggesting an impaired LCHAD step in long-chain FAO. LCHADD mice have progressively decreased visual performance and increased RPE degeneration starting at 6 months. LCHADD RPE have an altered structure and a two-fold increase in macrophages in the subretinal space. Finally, LCHADD RPE/sclera have differentially expressed genes compared to WT, including downregulation of genes important for RPE function and angiogenesis. Conclusions Overall, this LCHADD mouse model recapitulates early-stage chorioretinopathy seen in patients with LCHADD and is a useful model for studying LCHADD chorioretinopathy.
Collapse
Affiliation(s)
- Shannon J. Babcock
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, United States
| | - Allison G. Curtis
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Garen Gaston
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, United States
| | - Gabriela Elizondo
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, United States
| | - Melanie B. Gillingham
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, United States
| | - Renee C. Ryals
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, United States
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| |
Collapse
|
7
|
Elizondo G, Saini A, Gonzalez de Alba C, Gregor A, Harding CO, Gillingham MB, Vinocur JM. Cardiac phenotype in adolescents and young adults with long-chain 3-hydroxyacyl CoA dehydrogenase (LCHAD) deficiency. Genet Med 2024; 26:101123. [PMID: 38501492 DOI: 10.1016/j.gim.2024.101123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 03/20/2024] Open
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.
Collapse
Affiliation(s)
- Gabriela Elizondo
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR
| | - Ajesh Saini
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR; Portland State University, Urban Honors College, Portland, OR
| | | | - Ashley Gregor
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR
| | - Cary O Harding
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR
| | - Melanie B Gillingham
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR.
| | - Jeffrey M Vinocur
- Division of Pediatric Cardiology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
8
|
Mütze U, Ottenberger A, Gleich F, Maier EM, Lindner M, Husain RA, Palm K, Beblo S, Freisinger P, Santer R, Thimm E, vom Dahl S, Weinhold N, Grohmann‐Held K, Haase C, Hennermann JB, Hörbe‐Blindt A, Kamrath C, Marquardt I, Marquardt T, Behne R, Haas D, Spiekerkoetter U, Hoffmann GF, Garbade SF, Grünert SC, Kölker S. Neurological outcome in long-chain hydroxy fatty acid oxidation disorders. Ann Clin Transl Neurol 2024; 11:883-898. [PMID: 38263760 PMCID: PMC11021608 DOI: 10.1002/acn3.52002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/04/2024] [Indexed: 01/25/2024] Open
Abstract
OBJECTIVE This study aims to elucidate the long-term benefit of newborn screening (NBS) for individuals with long-chain 3-hydroxy-acyl-CoA dehydrogenase (LCHAD) and mitochondrial trifunctional protein (MTP) deficiency, inherited metabolic diseases included in NBS programs worldwide. METHODS German national multicenter study of individuals with confirmed LCHAD/MTP deficiency identified by NBS between 1999 and 2020 or selective metabolic screening. Analyses focused on NBS results, confirmatory diagnostics, and long-term clinical outcomes. RESULTS Sixty-seven individuals with LCHAD/MTP deficiency were included in the study, thereof 54 identified by NBS. All screened individuals with LCHAD deficiency survived, but four with MTP deficiency (14.8%) died during the study period. Despite NBS and early treatment neonatal decompensations (28%), symptomatic disease course (94%), later metabolic decompensations (80%), cardiomyopathy (28%), myopathy (82%), hepatopathy (32%), retinopathy (17%), and/or neuropathy (22%) occurred. Hospitalization rates were high (up to a mean of 2.4 times/year). Disease courses in screened individuals with LCHAD and MTP deficiency were similar except for neuropathy, occurring earlier in individuals with MTP deficiency (median 3.9 vs. 11.4 years; p = 0.0447). Achievement of dietary goals decreased with age, from 75% in the first year of life to 12% at age 10, and consensus group recommendations on dietary management were often not achieved. INTERPRETATION While NBS and early treatment result in improved (neonatal) survival, they cannot reliably prevent long-term morbidity in screened individuals with LCHAD/MTP deficiency, highlighting the urgent need of better therapeutic strategies and the development of disease course-altering treatment.
Collapse
Affiliation(s)
- Ulrike Mütze
- Medical Faculty of Heidelberg, Center for Child and Adolescent Medicine, Division of Child Neurology and Metabolic MedicineHeidelberg UniversityHeidelbergGermany
| | - Alina Ottenberger
- Medical Faculty of Heidelberg, Center for Child and Adolescent Medicine, Division of Child Neurology and Metabolic MedicineHeidelberg UniversityHeidelbergGermany
| | - Florian Gleich
- Medical Faculty of Heidelberg, Center for Child and Adolescent Medicine, Division of Child Neurology and Metabolic MedicineHeidelberg UniversityHeidelbergGermany
| | - Esther M. Maier
- Dr. von Hauner Children's Hospital, Ludwig‐Maximilians‐UniversityMunichGermany
| | - Martin Lindner
- Division of Pediatric NeurologyUniversity Children's Hospital FrankfurtFrankfurtGermany
| | - Ralf A. Husain
- Center for Inborn Metabolic Disorders, Department of NeuropediatricsJena University HospitalJenaGermany
| | - Katja Palm
- Division of Endocrinology, Diabetology and Metabolic MedicineUniversity Children's HospitalMagdeburgGermany
| | - Skadi Beblo
- Department of Women and Child Health, Hospital for Children and Adolescents, Center for Pediatric Research Leipzig (CPL)University Hospitals, University of LeipzigLeipzigGermany
| | - Peter Freisinger
- Children's Hospital Reutlingen, Klinikum am SteinenbergReutlingenGermany
| | - René Santer
- University Medical Center Hamburg‐Eppendorf, University Children's HospitalHamburgGermany
| | - Eva Thimm
- Department of General Pediatrics, Neonatology, and Pediatric CardiologyUniversity Children's Hospital, Heinrich Heine University DüsseldorfDüsseldorfGermany
| | - Stephan vom Dahl
- Department of Gastroenterology, Hepatology and Infectious DiseasesUniversity Hospital, Heinrich Heine University DüsseldorfDüsseldorfGermany
| | - Natalie Weinhold
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Center of Chronically Sick ChildrenCharité ‐ Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Karina Grohmann‐Held
- Department of Pediatrics and Adolescent MedicineUniversity Medicine GreifswaldGreifswaldGermany
| | - Claudia Haase
- Department of Pediatrics and Adolescent MedicineHelios Hospital ErfurtErfurtGermany
| | - Julia B. Hennermann
- Villa Metabolica, Center for Pediatric and Adolescent MedicineMainz University Medical CenterMainzGermany
| | | | - Clemens Kamrath
- Department of General Pediatrics and NeonatologyUniversity Hospital of Gießen and MarburgGießenGermany
| | - Iris Marquardt
- Department of Child NeurologyChildren's Hospital OldenburgOldenburgGermany
| | - Thorsten Marquardt
- Department of General Pediatrics, Metabolic DiseasesUniversity Children's Hospital MuensterMuensterGermany
| | - Robert Behne
- Medical Faculty of Heidelberg, Center for Child and Adolescent Medicine, Division of Child Neurology and Metabolic MedicineHeidelberg UniversityHeidelbergGermany
| | - Dorothea Haas
- Medical Faculty of Heidelberg, Center for Child and Adolescent Medicine, Division of Child Neurology and Metabolic MedicineHeidelberg UniversityHeidelbergGermany
| | - Ute Spiekerkoetter
- Department of General Pediatrics, Adolescent Medicine and NeonatologyMedical Center ‐ University of Freiburg, Faculty of MedicineFreiburgGermany
| | - Georg F. Hoffmann
- Medical Faculty of Heidelberg, Center for Child and Adolescent Medicine, Division of Child Neurology and Metabolic MedicineHeidelberg UniversityHeidelbergGermany
| | - Sven F. Garbade
- Medical Faculty of Heidelberg, Center for Child and Adolescent Medicine, Division of Child Neurology and Metabolic MedicineHeidelberg UniversityHeidelbergGermany
| | - Sarah C. Grünert
- Department of General Pediatrics, Adolescent Medicine and NeonatologyMedical Center ‐ University of Freiburg, Faculty of MedicineFreiburgGermany
| | - Stefan Kölker
- Medical Faculty of Heidelberg, Center for Child and Adolescent Medicine, Division of Child Neurology and Metabolic MedicineHeidelberg UniversityHeidelbergGermany
| |
Collapse
|
9
|
Wongchaisuwat N, Gillingham MB, Yang P, Everett L, Gregor A, Harding CO, Sahel JA, Nischal KK, Scanga HL, Black D, Vockley J, Arnold G, Pennesi ME. A proposal for an updated staging system for LCHADD retinopathy. Ophthalmic Genet 2024; 45:140-146. [PMID: 38288966 PMCID: PMC11010772 DOI: 10.1080/13816810.2024.2303682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 01/05/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVE To develop an updated staging system for long-chain 3-hydroxyacyl coenzyme A dehydrogenase deficiency (LCHADD) chorioretinopathy based on contemporary multimodal imaging and electrophysiology. METHODS We evaluated forty cases of patients with genetically confirmed LCHADD or trifunctional protein deficiency (TFPD) enrolled in a prospective natural history study. Wide-field fundus photographs, fundus autofluorescence (FAF), optical coherence tomography (OCT), and full-field electroretinogram (ffERG) were reviewed and graded for severity. RESULTS Two independent experts first graded fundus photos and electrophysiology to classify the stage of chorioretinopathy based upon an existing published system. With newer imaging modalities and improved electrophysiology, many patients did not fit cleanly into a single traditional staging group. Therefore, we developed a novel staging system that better delineated the progression of LCHADD retinopathy. We maintained the four previous delineated stages but created substages A and B in stages 2 to 3 to achieve better differentiation. DISCUSSION Previous staging systems of LCHADD chorioretinopathy relied on only on the assessment of standard 30 to 45-degree fundus photographs, visual acuity, fluorescein angiography (FA), and ffERG. Advances in recordings of ffERG and multimodal imaging with wider fields of view, allow better assessment of retinal changes. Following these advanced assessments, seven patients did not fit neatly into the original classification system and were therefore recategorized under the new proposed system. CONCLUSION The new proposed staging system improves the classification of LCHADD chorioretinopathy, with the potential to lead to a deeper understanding of the disease's progression and serve as a more reliable reference point for future therapeutic research.
Collapse
Affiliation(s)
- Nida Wongchaisuwat
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, Oregon, USA
- Department of Ophthalmology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Melanie B. Gillingham
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon, USA
| | - Paul Yang
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, Oregon, USA
| | - Lesley Everett
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, Oregon, USA
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon, USA
| | - Ashley Gregor
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon, USA
| | - Cary O. Harding
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon, USA
| | - Jose Alain Sahel
- Department of Ophthalmology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Ken K. Nischal
- Department of Ophthalmology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Hannah L. Scanga
- Department of Ophthalmology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Danielle Black
- Division of Genetic and Genomic Medicine, University of Pittsburgh Medical Center Children’s Hospital, Pittsburgh, Pennsylvania, USA
| | - Jerry Vockley
- Division of Genetic and Genomic Medicine, University of Pittsburgh Medical Center Children’s Hospital, Pittsburgh, Pennsylvania, USA
| | - Georgianne Arnold
- Division of Genetic and Genomic Medicine, University of Pittsburgh Medical Center Children’s Hospital, Pittsburgh, Pennsylvania, USA
| | - Mark E. Pennesi
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, Oregon, USA
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
10
|
Wongchaisuwat N, Wang J, Yang P, Everett L, Gregor A, Sahel JA, Nischal KK, Pennesi ME, Gillingham MB, Jia Y. Optical coherence tomography angiography of choroidal neovascularization in long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD). Am J Ophthalmol Case Rep 2023; 32:101958. [PMID: 38161518 PMCID: PMC10757195 DOI: 10.1016/j.ajoc.2023.101958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 10/26/2023] [Accepted: 11/02/2023] [Indexed: 01/03/2024] Open
Abstract
Purpose To report the clinical utility of optical coherence tomography angiography (OCTA) for demonstrating choroidal neovascularization (CNV) associated with Long-Chain 3-Hydroxyacyl-CoA Dehydrogenase Deficiency (LCHADD) retinopathy. Methods Thirty-three participants with LCHADD (age 7-36 years; median 17) were imaged with OCTA and the Center for Ophthalmic Optics & Lasers Angiography Reading Toolkit (COOL-ART) software was implemented to process OCTA scans. Results Seven participants (21 %; age 17-36 years; median 25) with LCHADD retinopathy demonstrated evidence of CNV by retinal examination or presence of CNV within outer retinal tissue on OCTA scans covering 3 × 3 and/or 6 × 6-mm. These sub-clinical CNVs are adjacent to hyperpigmented areas in the posterior pole. CNV presented at stage 2 or later of LCHADD retinopathy prior to the disappearance of RPE pigment in the macula. Conclusion OCTA can be applied as a non-invasive method to evaluate the retinal and choroidal microvasculature. OCTA can reveal CNV in LCHADD even when the clinical exam is inconclusive. These data suggest that the incidence of CNV is greater than expected and can occur even in the early stages of LCHADD retinopathy.
Collapse
Affiliation(s)
- Nida Wongchaisuwat
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, OR, USA
- Department of Ophthalmology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jie Wang
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, OR, USA
| | - Paul Yang
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, OR, USA
| | - Lesley Everett
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, OR, USA
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Ashley Gregor
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Jose Alain Sahel
- Vision Institute, University of Pittsburgh Medical Center and School of Medicine, Pennsylvania, USA
| | - Ken K. Nischal
- Vision Institute, University of Pittsburgh Medical Center and School of Medicine, Pennsylvania, USA
- UPMC Children's Hospital, Pennsylvania, USA
| | - Mark E. Pennesi
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, OR, USA
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Melanie B. Gillingham
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Yali Jia
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
11
|
Gaston G, Babcock S, Ryals R, Elizondo G, DeVine T, Wafai D, Packwood W, Holden S, Raber J, Lindner JR, Pennesi ME, Harding CO, Gillingham MB. A G1528C Hadha knock-in mouse model recapitulates aspects of human clinical phenotypes for long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency. Commun Biol 2023; 6:890. [PMID: 37644104 PMCID: PMC10465608 DOI: 10.1038/s42003-023-05268-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 08/21/2023] [Indexed: 08/31/2023] Open
Abstract
Long chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD) is a fatty acid oxidation disorder (FAOD) caused by a pathogenic variant, c.1528 G > C, in HADHA encoding the alpha subunit of trifunctional protein (TFPα). Individuals with LCHADD develop chorioretinopathy and peripheral neuropathy not observed in other FAODs in addition to the more ubiquitous symptoms of hypoketotic hypoglycemia, rhabdomyolysis and cardiomyopathy. We report a CRISPR/Cas9 generated knock-in murine model of G1528C in Hadha that recapitulates aspects of the human LCHADD phenotype. Homozygous pups are less numerous than expected from Mendelian probability, but survivors exhibit similar viability with wildtype (WT) littermates. Tissues of LCHADD homozygotes express TFPα protein, but LCHADD mice oxidize less fat and accumulate plasma 3-hydroxyacylcarnitines compared to WT mice. LCHADD mice exhibit lower ketones with fasting, exhaust earlier during treadmill exercise and develop a dilated cardiomyopathy compared to WT mice. In addition, LCHADD mice exhibit decreased visual performance, decreased cone function, and disruption of retinal pigment epithelium. Neurological function is affected, with impaired motor function during wire hang test and reduced open field activity. The G1528C knock-in mouse exhibits a phenotype similar to that observed in human patients; this model will be useful to explore pathophysiology and treatments for LCHADD in the future.
Collapse
Affiliation(s)
- Garen Gaston
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Shannon Babcock
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Renee Ryals
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
- Casey Eye Institute, Oregon Health and Science University, Portland, OR, USA
| | - Gabriela Elizondo
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Tiffany DeVine
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Dahlia Wafai
- Casey Eye Institute, Oregon Health and Science University, Portland, OR, USA
| | - William Packwood
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | - Sarah Holden
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, USA
| | - Jacob Raber
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, USA
- Departments of Neurology and Radiation Medicine, Oregon Health and Science University, Portland, OR, USA
- Division of Neuroscience, Oregon National Primate Research Center (ONPRC), Oregon Health and Science University, Portland, OR, USA
| | - Jonathan R Lindner
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
- Cardiovascular Division, University of Virginia Medical Center, Charlottesville, VA, USA
| | - Mark E Pennesi
- Casey Eye Institute, Oregon Health and Science University, Portland, OR, USA
| | - Cary O Harding
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Melanie B Gillingham
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
12
|
Successful management of rhabdomyolysis with triheptanoin in a child with severe long-chain 3-hydroxyacyl-coenzyme A dehydrogenase (LCHAD) deficiency. Neuromuscul Disord 2023; 33:315-318. [PMID: 36893607 DOI: 10.1016/j.nmd.2023.02.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 02/04/2023] [Accepted: 02/15/2023] [Indexed: 02/21/2023]
Abstract
Early-onset long-chain 3-hydroxyacyl-coenzyme A dehydrogenase (LCHAD) deficiency is a fatty acid β-oxidation disorder with a poor prognosis. Triheptanoin, an anaplerotic oil with odd-chain fatty acids can improve the disease course. The female patient presented here was diagnosed at the age of 4 months, and treatment was started as fat restriction, frequent feeding, and standard medium-chain triglyceride supplementation. In follow-up, she had frequent rhabdomyolysis episodes (∼8 per year). At the age of six, she had 13 episodes in 6 months, and triheptanoin was started as part of a compassionate use program. Following unrelated hospital stays due to multisystem inflammatory syndrome in children and a bloodstream infection, she had only 3 rhabdomyolysis episodes, and hospitalized days decreased from 73 to 11 during her first year with triheptanoin. Triheptanoin drastically decreased the frequency and severity of rhabdomyolysis, but progression of retinopathy was not altered.
Collapse
|
13
|
Baker JJ, Burton BK. Diagnosis and Clinical Management of Long-chain Fatty-acid Oxidation Disorders: A Review. TOUCHREVIEWS IN ENDOCRINOLOGY 2022; 17:108-111. [PMID: 35118456 DOI: 10.17925/ee.2021.17.2.108] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/13/2021] [Indexed: 11/24/2022]
Abstract
Long-chain fatty-acid oxidation disorders (LC-FAODs) are autosomal recessive inherited metabolic conditions that occur due to a disruption in the body's ability to perform mitochondrial beta oxidation. Expanded newborn screening is widening phenotypic understanding of these disorders, as well improving our knowledge of disease incidence. Management of these disorders is focused on avoidance of fasting, dietary changes and supplementation with energy sources that bypass the metabolic block. Recent US Food and Drug Administration approval of triheptanoin has improved the outcome for affected individuals. New research into dietary modifications and novel pharmacologic therapies continues for these disorders. In this article, we review the major LC-FAODs and their clinical presentation.
Collapse
Affiliation(s)
- Joshua J Baker
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA.,Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Barbara K Burton
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA.,Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
14
|
Sacconi R, Bandello F, Querques G. CHOROIDAL NEOVASCULARIZATION ASSOCIATED WITH LONG-CHAIN 3-HYDROXYACYL-CoA DEHYDROGENASE DEFICIENCY. Retin Cases Brief Rep 2022; 16:99-101. [PMID: 31479012 DOI: 10.1097/icb.0000000000000923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE To report the first case describing choroidal neovascularization in long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD) deficiency. METHODS Case report including multimodal imaging discussion. RESULTS A 21-year-old woman affected by LCHAD deficiency (confirmed by 1528 G>C homozygous mutation) was referred to our department for progressive visual decline in both eyes. Best-corrected visual acuities were 20/40 and 20/1,000 in the right and left eye, respectively. Ultra-widefield imaging, fluorescein angiography, structural optical coherence tomography, and optical coherence tomography angiography revealed the presence of macular and midperipheral chorioretinal atrophy complicated by a choroidal neovascularization in the left eye. CONCLUSION Ocular changes in LCHAD deficiency are long-term complications and severely affect the quality of life of patients. We report for the first time the evidence that choroidal neovascularization could complicate ocular changes accelerating the progressive vision impairment.
Collapse
Affiliation(s)
- Riccardo Sacconi
- Department of Ophthalmology, University Vita-Salute, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | | |
Collapse
|
15
|
García García LC, Zamorano Martín F, Rocha de Lossada C, García Lorente M, Luque Aranda G, Escudero Gómez J. Retinitis pigmentosa as a clinical presentation of LCHAD deficiency: A clinical case and review of the literature. ARCHIVOS DE LA SOCIEDAD ESPAÑOLA DE OFTALMOLOGÍA 2021; 96:496-499. [PMID: 34479707 DOI: 10.1016/j.oftale.2020.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/09/2020] [Indexed: 11/16/2022]
Abstract
Long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency is a rare metabolic disease caused by a specific mutation in the HADHA gene, which leads to an alteration in the metabolic pathway of fatty acids. Its most frequent form of presentation at the ophthalmological level is retinitis pigmentosa, and in some cases the ophthalmologist could be the first one to alert the other paediatric specialties to carry out a multidisciplinary approach to the case. The case is presented of a patient with long-chain 3-hydroxyacyl-CoA dehydrogenase deficit detected in neonatal screening, and which clinically debuted as pigmentary retinosis with no alteration in visual acuity as observed in the fundus images and optical coherence tomography of the retina provided. Finally, a review of the literature of this potentially lethal pathology is presented, and the main pathological and clinical features are highlighted.
Collapse
Affiliation(s)
- L C García García
- Servicio de Oftalmología, Hospital Regional Universitario de Málaga, Málaga, Spain; Servicio de Oftalmología, Hospital Universitario de Torrevieja, Torrevieja, Alicante, Spain.
| | - F Zamorano Martín
- Servicio de Oftalmología, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - C Rocha de Lossada
- Servicio de Oftalmología, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - M García Lorente
- Servicio de Oftalmología, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - G Luque Aranda
- Servicio de Oftalmología, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - J Escudero Gómez
- Servicio de Oftalmología, Hospital Regional Universitario de Málaga, Málaga, Spain
| |
Collapse
|
16
|
Rigaudière F, Delouvrier E, Le Gargasson JF, Milani P, Ogier de Baulny H, Schiff M. Long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD) deficiency and progressive retinopathy: one case report followed by ERGs, VEPs, EOG over a 17-year period. Doc Ophthalmol 2021; 142:371-380. [PMID: 33392894 DOI: 10.1007/s10633-020-09802-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 10/27/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND LCHAD (long-chain 3-hydroxyacyl-CoA dehydrogenase) deficiency is a rare genetic disorder of mitochondrial long-chain fatty acid oxidation inherited as a recessive trait. Affected patients can present with hypoglycaemia, rhabdomyolysis and cardiomyopathy. About half of the patients may suffer from retinopathy. CASE REPORT A 19-year-old girl was diagnosed as suffering from LCHAD deficiency with recurrent rhabdomyolysis episodes at the age of 7 months by an inaugural coma with hypoglycaemia and hepatomegaly. Appropriate dietary management with carnitine supplementation was initiated. Retinopathy was diagnosed at age two. Ophthalmological assessments including visual acuity, visual field, OCT, flash ERGs, P-ERG, flash VEPs and EOG recordings were conducted over a 17-year period. RESULTS Visual acuity was decreased. Fundi showed a progressive retinopathy and chorioretinopathy. Photophobia was noticed 2 years before the decrease in photopic-ERG amplitude with normal scotopic-ERGs. Scotopic-ERG amplitude decreased 10 years after the decrease in photopic-ERG amplitude. No EOG light rise was observed. Flash VEPs remained normal. These results suggest that the cone system dysfunction occurs largely prior to the rod system dysfunction with a relative preservation of the macula function. COMMENTS This dysfunction of cones prior to the dysfunction of rods was not reported previously. This could be related to mitochondrial energy failure in cones as cones are greater consumers of ATP than rods. This hypothesis needs to be further confirmed as other long-chain fatty oxidation defective patients (VLCAD and CPT2 deficiencies) do not exhibit retinopathy.
Collapse
Affiliation(s)
- Florence Rigaudière
- Service de Physiologie Clinique, Exploration Fonctionnelle, Hôpital Lariboisière, AP-HP, Paris, France. .,Faculté de Médecine Paris-Diderot, Université de Paris, Paris, France.
| | | | - Jean-François Le Gargasson
- Service de Physiologie Clinique, Exploration Fonctionnelle, Hôpital Lariboisière, AP-HP, Paris, France.,Faculté de Médecine Paris-Diderot, Université de Paris, Paris, France
| | - Paolo Milani
- Service de Physiologie Clinique, Exploration Fonctionnelle, Hôpital Lariboisière, AP-HP, Paris, France
| | - Hélène Ogier de Baulny
- Faculté de Médecine Paris-Diderot, Université de Paris, Paris, France.,Reference Center for Inborn Errors of Metabolism, Robert Debré Hospital, AP-HP, Paris, France
| | - Manuel Schiff
- Reference Center for Inborn Errors of Metabolism, Robert Debré Hospital, AP-HP, Paris, France.,Reference Center for Inborn Errors of Metabolism, Faculté de Médecine Paris-Descartes, Necker University Hospital, AP-HP, Université de Paris, Paris, France.,Institut Imagine, Inserm UMRS_1163, Paris, France
| |
Collapse
|
17
|
Dulz S, Atiskova Y, Engel P, Wildner J, Tsiakas K, Santer R. Retained visual function in a subset of patients with long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD). Ophthalmic Genet 2020; 42:23-27. [PMID: 33107778 DOI: 10.1080/13816810.2020.1836658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Introduction: LCHADD causes retinopathy associated with low vision, visual field defects, nyctalopia and myopia. We report a retrospective long-term single-center study of 6 LCHADD patients trying to clarify if early diagnosis has an impact on the course and outcome of chorioretinal degeneration. Methods: Long-term follow-up of visual acuity and staging of chorioretinal degeneration by fundus photography, optical coherence tomography (OCT) and autofluorescence (AF) in all six patients. Three patients (2 m/1 f; age 8-14.8 years) were diagnosed by newborn screening, a single patient early within the first year of life and treated promptly while the other two (1 m/1 f; age 23-24 years) were diagnosed later after developing symptoms. All carried HADHA variants; five were homozygous for the common p.E510Q variant, in one from the symptomatically diagnosed group p.[E510Q]; [R291*] was detected. Results: All patients showed retinal alterations, but early diagnosis was associated with a milder phenotype and a longer preservation of visual function. Among symptomatic patients, only one showed mild retinal involvement at the time of diagnosis. Conclusion: Despite the small number our study suggests that early diagnosis does not prevent retinopathy but might contribute to a milder phenotype with retained good visual acuity over time. OCT and AF are reliable non-invasive diagnostic tools to estimate the progression of early-stage retinal changes in LCHADD patients.
Collapse
Affiliation(s)
- Simon Dulz
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf , Hamburg, Germany.,University Children's Hospital, University Medical Center Hamburg-Eppendorf , Hamburg, Germany
| | - Yevgeniya Atiskova
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf , Hamburg, Germany.,University Children's Hospital, University Medical Center Hamburg-Eppendorf , Hamburg, Germany
| | - Peter Engel
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf , Hamburg, Germany.,University Children's Hospital, University Medical Center Hamburg-Eppendorf , Hamburg, Germany
| | - Jan Wildner
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf , Hamburg, Germany.,University Children's Hospital, University Medical Center Hamburg-Eppendorf , Hamburg, Germany
| | - Konstantinos Tsiakas
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf , Hamburg, Germany.,University Children's Hospital, University Medical Center Hamburg-Eppendorf , Hamburg, Germany
| | - Rene Santer
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf , Hamburg, Germany.,University Children's Hospital, University Medical Center Hamburg-Eppendorf , Hamburg, Germany
| |
Collapse
|
18
|
García García LC, Zamorano Martín F, Rocha de Lossada C, García Lorente M, Luque Aranda G, Escudero Gómez J. Retinitis pigmentosa as a clinical presentation of LCHAD deficiency: A clinical case and review of the literature. ACTA ACUST UNITED AC 2020. [PMID: 32943256 DOI: 10.1016/j.oftal.2020.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency is a rare metabolic disease caused by a specific mutation in the HADHA gene, which leads to an alteration in the metabolic pathway of fatty acids. Its most frequent form of presentation at the ophthalmological level is retinitis pigmentosa, and in some cases the ophthalmologist could be the first one to alert the other paediatric specialties to carry out a multidisciplinary approach to the case. The case is presented of a patient with long-chain 3-hydroxyacyl-CoA dehydrogenase deficit detected in neonatal screening, and which clinically debuted as pigmentary retinosis with no alteration in visual acuity as observed in the fundus images and optical coherence tomography of the retina provided. Finally, a review of the literature of this potentially lethal pathology is presented, and the main pathological and clinical features are highlighted.
Collapse
Affiliation(s)
- L C García García
- Servicio de Oftalmología, Hospital Regional Universitario de Málaga, Málaga, España; Servicio de Oftalmología, Hospital Universitario de Torrevieja, Torrevieja, Alicante, España.
| | - F Zamorano Martín
- Servicio de Oftalmología, Hospital Regional Universitario de Málaga, Málaga, España
| | - C Rocha de Lossada
- Servicio de Oftalmología, Hospital Regional Universitario de Málaga, Málaga, España
| | - M García Lorente
- Servicio de Oftalmología, Hospital Regional Universitario de Málaga, Málaga, España
| | - G Luque Aranda
- Servicio de Oftalmología, Hospital Regional Universitario de Málaga, Málaga, España
| | - J Escudero Gómez
- Servicio de Oftalmología, Hospital Regional Universitario de Málaga, Málaga, España
| |
Collapse
|
19
|
Elizondo G, Matern D, Vockley J, Harding CO, Gillingham MB. Effects of fasting, feeding and exercise on plasma acylcarnitines among subjects with CPT2D, VLCADD and LCHADD/TFPD. Mol Genet Metab 2020; 131:90-97. [PMID: 32928639 PMCID: PMC8048763 DOI: 10.1016/j.ymgme.2020.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 11/16/2022]
Abstract
BACKGROUND The plasma acylcarnitine profile is frequently used as a biochemical assessment for follow-up in diagnosed patients with fatty acid oxidation disorders (FAODs). Disease specific acylcarnitine species are elevated during metabolic decompensation but there is clinical and biochemical heterogeneity among patients and limited data on the utility of an acylcarnitine profile for routine clinical monitoring. METHODS We evaluated plasma acylcarnitine profiles from 30 diagnosed patients with long-chain FAODs (carnitine palmitoyltransferase-2 (CPT2), very long-chain acyl-CoA dehydrogenase (VLCAD), and long-chain 3-hydroxy acyl-CoA dehydrogenase or mitochondrial trifunctional protein (LCHAD/TFP) deficiencies) collected after an overnight fast, after feeding a controlled low-fat diet, and before and after moderate exercise. Our purpose was to describe the variability in this biomarker and how various physiologic states effect the acylcarnitine concentrations in circulation. RESULTS Disease specific acylcarnitine species were higher after an overnight fast and decreased by approximately 60% two hours after a controlled breakfast meal. Moderate-intensity exercise increased the acylcarnitine species but it varied by diagnosis. When analyzed for a genotype/phenotype correlation, the presence of the common LCHADD mutation (c.1528G > C) was associated with higher levels of 3-hydroxyacylcarnitines than in patients with other mutations. CONCLUSIONS We found that feeding consistently suppressed and that moderate intensity exercise increased disease specific acylcarnitine species, but the response to exercise was highly variable across subjects and diagnoses. The clinical utility of routine plasma acylcarnitine analysis for outpatient treatment monitoring remains questionable; however, if acylcarnitine profiles are measured in the clinical setting, standardized procedures are required for sample collection to be of value.
Collapse
Affiliation(s)
- Gabriela Elizondo
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Dietrich Matern
- Biochemical Genetics Laboratory, Mayo Clinic, Rochester, MN, United States of America
| | - Jerry Vockley
- Department of Pediatrics University of Pittsburgh School of Medicine, Center for Rare Disease Therapy, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, United States of America
| | - Cary O Harding
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Melanie B Gillingham
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, United States of America; Biochemical Genetics Laboratory, Mayo Clinic, Rochester, MN, United States of America.
| |
Collapse
|
20
|
Merritt JL, Norris M, Kanungo S. Fatty acid oxidation disorders. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:473. [PMID: 30740404 DOI: 10.21037/atm.2018.10.57] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Fatty acid oxidation disorders (FAODs) are inborn errors of metabolism due to disruption of either mitochondrial β-oxidation or the fatty acid transport using the carnitine transport pathway. The presentation of a FAOD will depend upon the specific disorder, but common elements may be seen, and ultimately require a similar treatment. Initial presentations of the FAODs in the neonatal period with severe symptoms include cardiomyopathy, while during infancy and childhood liver dysfunction and hypoketotic hypoglycemia are common. Episodic rhabdomyolysis is frequently the initial presentation during or after adolescence; although, these symptoms may develop at any age for most of the FAODs The treatment of all FAOD's include avoidance of fasting, aggressive treatment during illness, and supplementation of carnitine, if necessary. The long-chain FAODs differ by requiring a fat-restricted diet and supplementation of medium chain triglyceride oil and often docosahexaenoic acid (DHA)-an essential fatty acid, crucial for brain, visual, and immune functions and prevention of fat soluble vitamin deficiencies. The FAOD are a group of autosomal recessive disorders associated with significant morbidity and mortality, but early diagnosis on newborn screening (NBS) and early initiation of treatment are improving outcomes. There is a need for clinical studies including randomized, controlled, therapeutic trials to continue to evaluate current understanding and to implement future therapies.
Collapse
Affiliation(s)
- J Lawrence Merritt
- Department of Pediatrics, University of Washington, Seattle, WA, USA.,Biochemial Genetics, Seattle Children's Hospital, Seattle, WA, USA
| | - Marie Norris
- Biochemial Genetics, Seattle Children's Hospital, Seattle, WA, USA
| | - Shibani Kanungo
- Department of Pediatrics and Adolescent Medicine, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA
| |
Collapse
|
21
|
Karlstetter M, Dannhausen K, Langmann T. Mikroglia und Immuntherapien bei degenerativen Netzhauterkrankungen. MED GENET-BERLIN 2017. [DOI: 10.1007/s11825-017-0132-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Zusammenfassung
Bei allen bisher im Detail untersuchten erblichen Netzhautdegenerationen liegt eine dem Erkrankungsverlauf abträgliche chronische Aktivierung des angeborenen Immunsystems zugrunde. Vor allem residente Mikrogliazellen der Netzhaut und verschiedene Proteine des löslichen Komplementsystems tragen zu einer Schädigung von Photorezeptoren und retinalem Pigmentepithel bei. Sowohl spezifische Zielstrukturen auf reaktiven Immunzellen als auch fehlregulierte lösliche Immunmodulatoren bieten neue Ansatzpunkte für Therapien, um das Überleben der Netzhaut trotz genetischer Prädisposition zur Degeneration zu fördern. Dieser Beitrag gibt Einblick in die wesentlichen Regulationsmechanismen der Netzhautimmunologie, diskutiert die mögliche Verwendung immunologischer Biomarker für die Netzhautdiagnostik und zeigt immunmodulierende Therapieansätze durch Biologika und endogene Botenstoffe auf.
Collapse
Affiliation(s)
- Marcus Karlstetter
- Aff1 0000 0000 8852 305X grid.411097.a Lehrstuhl für Experimentelle Immunologie des Auges, Zentrum für Augenheilkunde Uniklinik Köln Joseph-Stelzmann-Str. 9 50931 Köln Deutschland
| | - Katharina Dannhausen
- Aff1 0000 0000 8852 305X grid.411097.a Lehrstuhl für Experimentelle Immunologie des Auges, Zentrum für Augenheilkunde Uniklinik Köln Joseph-Stelzmann-Str. 9 50931 Köln Deutschland
| | - Thomas Langmann
- Aff1 0000 0000 8852 305X grid.411097.a Lehrstuhl für Experimentelle Immunologie des Auges, Zentrum für Augenheilkunde Uniklinik Köln Joseph-Stelzmann-Str. 9 50931 Köln Deutschland
| |
Collapse
|
22
|
Fahnehjelm KT, Liu Y, Olsson D, Amrén U, Haglind CB, Holmström G, Halldin M, Andreasson S, Nordenström A. Most patients with long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency develop pathological or subnormal retinal function. Acta Paediatr 2016; 105:1451-1460. [PMID: 27461099 DOI: 10.1111/apa.13536] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 07/25/2016] [Indexed: 12/27/2022]
Abstract
AIM There have been few studies on long-term electroretinographic findings in patients with long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD). This study correlated long-term electroretinographic findings with age, metabolic control and clinical symptoms. METHODS We examined 12 Swedish patients with LCHADD. Visual acuity testing, fundus examinations, optical coherence tomography and electroretinography were performed. The results were correlated to age, the levels of 3-hydroxyacylcarnitine and acylcarnitine and clinical metabolic control. RESULTS Blindness or moderate visual impairment was found in two patients. Retinal pigmentation, atrophy and fibrosis were present in 11, seven and one of the patients, respectively, and optical coherence tomography showed retinal thinning in three of the six patients examined. Electroretinography was performed on 11 of the 12 patients. It was pathological, with reduced rod and cone responses, in five patients, subnormal in four and was related to poor clinical metabolic control and severe neonatal symptoms. Repeated electroretinographies revealed reduced function with increasing age. CONCLUSION More than 80% of the LCHADD patients developed pathological or subnormal retinal function. This was more pronounced in patients with neonatal symptoms, but ameliorated by strict dietary treatment. Annual ophthalmological follow-ups, with electroretinography every second or third year, are recommended.
Collapse
Affiliation(s)
- Kristina Teär Fahnehjelm
- Department of Clinical Neuroscience; Karolinska Institutet; Stockholm Sweden
- St Erik Eye Hospital; Stockholm Sweden
- Institute of Neuroscience and Physiology; Sahlgrenska Akademin; University of Gothenburg; Gothenburg Sweden
| | - Ying Liu
- Department of Clinical Neurophysiology; Karolinska University Hospital; Huddinge Sweden
- Department of Ophthalmology; The South Hospital; Stockholm Sweden
| | - David Olsson
- Department of Women's and Children's Health; Karolinska Institutet; Stockholm Sweden
- Department of Paediatrics; Karolinska University Hospital; Stockholm Sweden
| | - Urban Amrén
- Department of Clinical Neuroscience; Karolinska Institutet; Stockholm Sweden
- St Erik Eye Hospital; Stockholm Sweden
| | - Charlotte Bieneck Haglind
- Department of Women's and Children's Health; Karolinska Institutet; Stockholm Sweden
- Department of Paediatrics; Karolinska University Hospital; Stockholm Sweden
| | - Gerd Holmström
- Department of Neuroscience/ophthalmology; Uppsala University Hospital; Uppsala Sweden
| | - Maria Halldin
- Department of Paediatric endocrinology; Uppsala University Children's Hospital; Uppsala Sweden
| | - Sten Andreasson
- Department of Ophthalmology; University of Lund; Lund Sweden
| | - Anna Nordenström
- Department of Women's and Children's Health; Karolinska Institutet; Stockholm Sweden
- Department of Paediatrics; Karolinska University Hospital; Stockholm Sweden
| |
Collapse
|
23
|
Boese EA, Jain N, Jia Y, Schlechter CL, Harding CO, Gao SS, Patel RC, Huang D, Weleber RG, Gillingham MB, Pennesi ME. Characterization of Chorioretinopathy Associated with Mitochondrial Trifunctional Protein Disorders: Long-Term Follow-up of 21 Cases. Ophthalmology 2016; 123:2183-95. [PMID: 27491397 PMCID: PMC5035590 DOI: 10.1016/j.ophtha.2016.06.048] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 06/11/2016] [Accepted: 06/16/2016] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To assess long-term effects of genotype on chorioretinopathy severity in patients with mitochondrial trifunctional protein (MTP) disorders. DESIGN Retrospective case series. PARTICIPANTS Consecutive patients with MTP disorders evaluated at a single center from 1994 through 2015, including 18 patients with long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD) and 3 patients with trifunctional protein deficiency (TFPD). METHODS Local records from all visits were reviewed. Every participant underwent a complete ophthalmic examination and was evaluated by a metabolic physician and dietitian. Nine patients underwent ancillary funduscopic imaging including optical coherence tomography (OCT) and OCT angiography. MAIN OUTCOME MEASURES The primary outcome measure was best-corrected visual acuity at the final visit. Secondary outcome measures included spherical equivalent refraction, visual fields, electroretinography B-wave amplitudes, and qualitative imaging findings. RESULTS Participants were followed up for a median of 5.6 years (range 0.3-20.2 years). The median age of LCHADD participants at initial and final visits was 2.3 and 11.9 years, whereas that for TFPD participants at initial and final visits was 4.7 and 15.5 years, respectively. Four long-term survivors older than 16 years were included (3 with LCHADD and 1 with TFPD). The LCHADD participants demonstrated a steady decline in visual acuity from an average of 0.23 logarithm of the minimum angle of resolution (logMAR; Snellen equivalent, 20/34) at baseline to 0.42 logMAR (Snellen equivalent, 20/53) at the final visit, whereas TFPD patients maintained excellent acuity throughout follow-up. Participants with LCHADD, but not TFPD, showed an increasing myopia with a mean decrease in spherical equivalent refraction of 0.24 diopters per year. Visual fields showed sensitivity losses centrally associated with defects on OCT. Multimodal imaging demonstrated progressive atrophy of the outer retina in LCHADD, often preceded by the formation of outer retinal tubulations and choriocapillaris dropout. Electroretinography findings support the more severe clinical profile of LCHADD patients compared with TFPD patients; the function of both rods and cones are attenuated diffusely in LCHADD patients, but are within normal limits for TFPD patients. CONCLUSIONS Despite improved survival with early diagnosis, medical management, and dietary treatment, participants with the LCHADD subtype of MTP disorder continue to demonstrate visually disabling chorioretinopathy. Multimodal imaging is most consistent with choriocapillaris loss exceeding photoreceptor loss.
Collapse
Affiliation(s)
- Erin A Boese
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon
| | - Nieraj Jain
- Department of Ophthalmology, Emory University, Atlanta, Georgia
| | - Yali Jia
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon
| | - Catie L Schlechter
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon
| | - Cary O Harding
- Molecular & Medical Genetics, Oregon Health & Science University, Portland, Oregon
| | - Simon S Gao
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon
| | - Rachel C Patel
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon
| | - David Huang
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon
| | - Richard G Weleber
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon
| | - Melanie B Gillingham
- Molecular & Medical Genetics, Oregon Health & Science University, Portland, Oregon
| | - Mark E Pennesi
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon.
| |
Collapse
|
24
|
MULTIMODAL IMAGING AND ELECTRORETINOGRAPHY IN LONG-CHAIN 3-HYDROXYACYL COENZYME A DEHYDROGENASE DEFICIENCY. Retin Cases Brief Rep 2016; 11 Suppl 1:S107-S112. [PMID: 27652820 DOI: 10.1097/icb.0000000000000428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE To report a case of pigmentary retinopathy in long-chain 3-hydroxyacyl coenzyme A dehydrogenase deficiency using multimodal imaging techniques. METHODS Case report. RESULTS An 8-year-old boy with a history of failure to thrive and a diagnosis of long-chain 3-hydroxyacyl coenzyme A dehydrogenase deficiency was referred for examination. Examination revealed a pigmentary retinopathy with macular atrophy; electroretinography results were consistent with a rod-cone dystrophy. Fundus autofluorescence and optical coherence tomography revealed retinal pigment epithelium atrophy. Follow-up examination findings showed increased severity of retinopathy on electroretinography, with optical coherence tomography angiography revealing enhanced visualization of choroidal vessels. CONCLUSION This report reveals that long-chain 3-hydroxyacyl coenzyme A dehydrogenase deficiency can be characterized as a progressive rod-cone dystrophy, with multi-modal imaging techniques used to describe this condition. In particular, optical coherence tomography angiography can be used to further characterize this condition.
Collapse
|
25
|
Immonen T, Turanlahti M, Paganus A, Keskinen P, Tyni T, Lapatto R. Earlier diagnosis and strict diets improve the survival rate and clinical course of long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency. Acta Paediatr 2016; 105:549-54. [PMID: 26676313 DOI: 10.1111/apa.13313] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 08/14/2015] [Accepted: 12/11/2015] [Indexed: 12/14/2022]
Abstract
AIM Long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD) is a severe metabolic disease that, without treatment, often leads to premature death or serious handicap. The aim of this study was to evaluate the clinical course of LCHADD with the homozygous 1528G>C (E510Q) mutation when patients underwent strict dietary treatment. METHODS From 1997 to 2010, 16 patients with LCHADD were diagnosed in Finland. They were followed up, and data were prospectively collected as they emerged. Clinical data before diagnosis were retrospectively collected from hospital records. This cohort was compared with an earlier cohort of patients diagnosed from 1976 to 1996. RESULTS The disease presented from birth to five months of age with failure to thrive, hypotonia, hepatomegaly, metabolic acidosis, cardiomyopathy and hypoketotic hypoglycaemia. In this cohort, the therapeutic delay was 0-30 days and the survival rate at the end of the study was 62.5% compared with 10-year survival rate of 14.3% for the earlier cohort. The survivors were in good overall condition, but some of them had developed mild retinopathy or mild neuropathy. CONCLUSION Earlier diagnosis and stricter dietary regimes improved the survival rates and clinical course of patients with LCHADD in Finland. However, improvements in therapy are still needed to prevent the development of long-term complications, such as retinopathy and neuropathy.
Collapse
Affiliation(s)
- Tuuli Immonen
- Children's Hospital; University of Helsinki and Helsinki University Hospital; Helsinki Finland
| | - Maila Turanlahti
- Children's Hospital; University of Helsinki and Helsinki University Hospital; Helsinki Finland
| | - Aila Paganus
- Children's Hospital; University of Helsinki and Helsinki University Hospital; Helsinki Finland
| | - Päivi Keskinen
- Pediatric Research Centre; University of Tampere; Tampere University Hospital; Tampere Finland
| | - Tiina Tyni
- Children's Hospital; University of Helsinki and Helsinki University Hospital; Helsinki Finland
| | - Risto Lapatto
- Children's Hospital; University of Helsinki and Helsinki University Hospital; Helsinki Finland
| |
Collapse
|
26
|
De Biase I, Viau KS, Liu A, Yuzyuk T, Botto LD, Pasquali M, Longo N. Diagnosis, Treatment, and Clinical Outcome of Patients with Mitochondrial Trifunctional Protein/Long-Chain 3-Hydroxy Acyl-CoA Dehydrogenase Deficiency. JIMD Rep 2016; 31:63-71. [PMID: 27117294 DOI: 10.1007/8904_2016_558] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 03/15/2016] [Accepted: 03/16/2016] [Indexed: 12/02/2022] Open
Abstract
Deficiency of the mitochondrial trifunctional protein (TFP) and long-chain 3-Hydroxy Acyl-CoA dehydrogenase (LCHAD) impairs long-chain fatty acid oxidation and presents with hypoglycemia, cardiac, liver, eye, and muscle involvement. Without treatment, both conditions can be life-threatening. These diseases are identified by newborn screening (NBS), but the impact of early treatment on long-term clinical outcome is unknown. Moreover, there is lack of consensus on treatment, particularly on the use of carnitine supplementation. Here, we report clinical and biochemical data in five patients with TFP/LCHAD deficiency, three of whom were diagnosed by newborn screening. All patients had signs and symptoms related to their metabolic disorder, including hypoglycemia, elevated creatine kinase (CK), and rhabdomyolysis, and experienced episodes of metabolic decompensation triggered by illness. Treatment was started shortly after diagnosis in all patients and consisted of a diet low in long-chain fats supplemented with medium chain triglycerides (MCT), essential fatty acids, and low-dose carnitine (25 mg/kg/day). Patients had growth restriction early in life that resolved after 2 years of age. All patients but the youngest (2 years old) developed pigmentary retinopathy. Long-chain hydroxylated acylcarnitines did not change significantly with age, but increased during acute illnesses. Free carnitine levels were maintained within the normal range and did not correlate with long-chain hydroxylated acylcarnitines. These results show that patients with LCHAD deficiency can have normal growth and development with appropriate treatment. Low-dose carnitine supplements prevented carnitine deficiency and did not result in increased long-chain hydroxylated acylcarnitines or any specific toxicity.
Collapse
Affiliation(s)
- Irene De Biase
- Department of Pathology, University of Utah, Salt Lake City, UT, 84108, USA. .,ARUP Laboratories, 500 Chipeta Way, Salt Lake City, UT, 84108, USA. .,ARUP Institute of Clinical and Experimental Pathology, Salt Lake City, UT, USA.
| | - Krista S Viau
- Department of Pediatrics, University of Utah, Salt Lake City, UT, 84108, USA
| | - Aiping Liu
- ARUP Institute of Clinical and Experimental Pathology, Salt Lake City, UT, USA
| | - Tatiana Yuzyuk
- Department of Pathology, University of Utah, Salt Lake City, UT, 84108, USA.,ARUP Laboratories, 500 Chipeta Way, Salt Lake City, UT, 84108, USA.,ARUP Institute of Clinical and Experimental Pathology, Salt Lake City, UT, USA
| | - Lorenzo D Botto
- Department of Pediatrics, University of Utah, Salt Lake City, UT, 84108, USA
| | - Marzia Pasquali
- Department of Pathology, University of Utah, Salt Lake City, UT, 84108, USA.,ARUP Laboratories, 500 Chipeta Way, Salt Lake City, UT, 84108, USA.,ARUP Institute of Clinical and Experimental Pathology, Salt Lake City, UT, USA.,Department of Pediatrics, University of Utah, Salt Lake City, UT, 84108, USA
| | - Nicola Longo
- Department of Pathology, University of Utah, Salt Lake City, UT, 84108, USA.,ARUP Laboratories, 500 Chipeta Way, Salt Lake City, UT, 84108, USA.,ARUP Institute of Clinical and Experimental Pathology, Salt Lake City, UT, USA.,Department of Pediatrics, University of Utah, Salt Lake City, UT, 84108, USA
| |
Collapse
|
27
|
Immonen T, Ahola E, Toppila J, Lapatto R, Tyni T, Lauronen L. Peripheral neuropathy in patients with long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency - A follow-up EMG study of 12 patients. Eur J Paediatr Neurol 2016; 20:38-44. [PMID: 26653362 DOI: 10.1016/j.ejpn.2015.10.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Revised: 10/14/2015] [Accepted: 10/21/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND The neonatal screening and early start of the dietary therapy have improved the outcome of long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD). The acute symptoms of LCHADD are hypoketotic hypoglycemia, failure to thrive, hepatopathy and rhabdomyolysis. Long term complications are retinopathy and neuropathy. Speculated etiology of these long term complications are the accumulation and toxicity of hydroxylacylcarnitines and long-chain fatty acid metabolites or deficiency of essential fatty acids. AIMS To study the possible development of polyneuropathy in LCHADD patients with current dietary regimen. METHODS Development of polyneuropathy in 12 LCHADD patients with the homozygous common mutation c.G1528C was evaluated with electroneurography (ENG) studies. The ENG was done 1-12 times to each patient, between the ages of 3 and 40 years. Clinical data of the patients were collected from the patient records. RESULTS The first sign of polyneuropathy was detected between the ages of 6-12 years, the first abnormality being reduction of the sensory amplitudes of the sural nerves. With time, progression was detected by abnormalities in sensory responses extending to upper limbs, as well as abnormalities in motor responses in lower limbs. Altogether, eight of the patients had polyneuropathy, despite good compliancy of the diet. CONCLUSIONS This study is the first to report the evolution of polyneuropathy with clinical neurophysiological methods in a relative large LCHADD patient group. Despite early start, and good compliance of the therapy, 6/10 of the younger patients developed neuropathy. However, in most patients the polyneuropathy was less severe than previously described.
Collapse
Affiliation(s)
- Tuuli Immonen
- Children's Hospital, University of Helsinki, Helsinki University Hospital, Finland.
| | - Emilia Ahola
- Children's Hospital, University of Helsinki, Helsinki University Hospital, Finland
| | - Jussi Toppila
- Department of Clinical Neurophysiology, Children's Hospital, University of Helsinki, HUS Medical Imaging Center, Finland
| | - Risto Lapatto
- Children's Hospital, University of Helsinki, Helsinki University Hospital, Finland
| | - Tiina Tyni
- Children's Hospital, University of Helsinki, Helsinki University Hospital, Finland
| | - Leena Lauronen
- Department of Clinical Neurophysiology, Children's Hospital, University of Helsinki, HUS Medical Imaging Center, Finland
| |
Collapse
|
28
|
Strandqvist A, Haglind CB, Zetterström RH, Nemeth A, von Döbeln U, Stenlid MH, Nordenström A. Neuropsychological Development in Patients with Long-Chain 3-Hydroxyacyl-CoA Dehydrogenase (LCHAD) Deficiency. JIMD Rep 2015; 28:75-84. [PMID: 26545880 DOI: 10.1007/8904_2015_505] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 09/24/2015] [Accepted: 09/28/2015] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Reports on cognitive outcomes in long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD) are scarce. We present results from neuropsychological assessments of eight patients diagnosed with LCHADD prior to newborn screening with regard to clinical disease severity. METHODS Intellectual ability and adaptive and executive functions were assessed using age-appropriate Wechsler Scales, Adaptive Behavior Assessment Scales (ABAS), and Behavior Rating Inventory of Executive Function (BRIEF). RESULTS Five patients performed in the normal range on IQ tests but with lower scores on verbal working memory. In addition, they had lower parent-rated adaptive and executive functions.Three patients had intellectual disabilities with IQs below normal and/or autism spectrum disorders. In addition, they had low results on parent-rated adaptive functions. (Two of these patients had epilepsy.) Conclusions: Patients with LCHADD seem to have a specific cognitive pattern, with presentation as intellectual disability and specific autistic deficiencies or a normal IQ with weaknesses in auditive verbal memory and adaptive and executive functions. Future studies are warranted to investigate whether newborn screening programs and early treatment may promote improved neuropsychological development and outcomes.
Collapse
Affiliation(s)
- A Strandqvist
- Department of Women and Children's Health, Karolinska Institutet, 171 76, Stockholm, Sweden.,Department of Psychology, Karolinska University Hospital, Stockholm, Sweden
| | - C Bieneck Haglind
- Department of Women and Children's Health, Karolinska Institutet, 171 76, Stockholm, Sweden. .,Department of Pediatrics, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden.
| | - R H Zetterström
- Departments of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - A Nemeth
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.,Department of Pediatric Gastroenterology, Hepatology and Nutrition, Karolinska University Hospital, Stockholm, Sweden
| | - U von Döbeln
- Center for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden.,Department of Laboratory Medicine, Division for Metabolic Diseases, Karolinska Institutet, Stockholm, Sweden
| | - M Halldin Stenlid
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - A Nordenström
- Department of Women and Children's Health, Karolinska Institutet, 171 76, Stockholm, Sweden.,Department of Pediatric Endocrinology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
29
|
Olpin SE, Murphy E, Kirk RJ, Taylor RW, Quinlivan R. The investigation and management of metabolic myopathies. J Clin Pathol 2015; 68:410-7. [DOI: 10.1136/jclinpath-2014-202808] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/25/2015] [Indexed: 01/19/2023]
Abstract
Metabolic myopathies (MM) are rare inherited primary muscle disorders that are mainly due to abnormalities of muscle energy metabolism resulting in skeletal muscle dysfunction. These diseases include disorders of fatty acid oxidation, glyco(geno)lytic muscle disorders and mitochondrial respiratory chain (MRC) disease. Clinically these disorders present with a range of symptoms including infantile hypotonia, myalgia/exercise tolerance, chronic or acute muscle weakness, cramps/spasms/stiffness or episodic acute rhabdomyolysis. The precipitant may be fasting, infection, general anaesthesia, heat/cold or most commonly, exercise. However, the differential diagnosis includes a wide range of both acquired and inherited conditions and these include exposure to drugs/toxins, inflammatory myopathies, dystrophies and channelopathies. Streamlining of existing diagnostic protocols has now become a realistic prospect given the availability of second-generation sequencing. A diagnostic pathway using a ‘rhabdomyolysis’ gene panel at an early stage of the diagnostic process is proposed. Following detailed clinical evaluation and first-line investigations, some patients will be identified as candidates for McArdle disease/glycogen storage disease type V or MRC disease and these will be referred directly to the specialised services. However, for the majority of patients, second-line investigation is best undertaken through next-generation sequencing using a ‘rhabdomyolysis’ gene panel. Following molecular analysis and careful evaluation of the findings, some patients will receive a clear diagnosis. Further functional or specific targeted testing may be required in other patients to evaluate the significance of uncertain/equivocal findings. For patients with no clear diagnosis, further investigations will be required through a specialist centre.
Collapse
|
30
|
Haglind CB, Nordenström A, Ask S, von Döbeln U, Gustafsson J, Stenlid MH. Increased and early lipolysis in children with long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD) deficiency during fast. J Inherit Metab Dis 2015; 38:315-22. [PMID: 25141826 DOI: 10.1007/s10545-014-9750-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 07/03/2014] [Accepted: 07/16/2014] [Indexed: 12/31/2022]
Abstract
Children with long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHAD) have a defect in the degradation of long-chain fatty acids and are at risk of hypoketotic hypoglycemia and insufficient energy production as well as accumulation of toxic fatty acid intermediates. Knowledge on substrate metabolism in children with LCHAD deficiency during fasting is limited. Treatment guidelines differ between centers, both as far as length of fasting periods and need for night feeds are concerned. To increase the understanding of fasting intolerance and improve treatment recommendations, children with LCHAD deficiency were investigated with stable isotope technique, microdialysis, and indirect calometry, in order to assess lipolysis and glucose production during 6 h of fasting. We found an early and increased lipolysis and accumulation of long chain acylcarnitines after 4 h of fasting, albeit no patients developed hypoglycemia. The rate of glycerol production, reflecting lipolysis, averaged 7.7 ± 1.6 µmol/kg/min, which is higher compared to that of peers. The rate of glucose production was normal for age; 19.6 ± 3.4 µmol/kg/min (3.5 ± 0.6 mg/kg/min). Resting energy expenditure was also normal, even though the respiratory quotient was increased indicating mainly glucose oxidation. The results show that lipolysis and accumulation of long chain acylcarnitines occurs before hypoglycemia in fasting children with LCHAD, which may indicate more limited fasting tolerance than previously suggested.
Collapse
Affiliation(s)
- C Bieneck Haglind
- Women's and Children's Health, Karolinska Institute, Stockholm, Sweden,
| | | | | | | | | | | |
Collapse
|
31
|
Karall D, Brunner-Krainz M, Kogelnig K, Konstantopoulou V, Maier EM, Möslinger D, Plecko B, Sperl W, Volkmar B, Scholl-Bürgi S. Clinical outcome, biochemical and therapeutic follow-up in 14 Austrian patients with Long-Chain 3-Hydroxy Acyl CoA Dehydrogenase Deficiency (LCHADD). Orphanet J Rare Dis 2015; 10:21. [PMID: 25888220 PMCID: PMC4407779 DOI: 10.1186/s13023-015-0236-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 01/29/2015] [Indexed: 12/31/2022] Open
Abstract
Background LCHADD is a long-fatty acid oxidation disorder with immediate symptoms and long-term complications. We evaluated data on clinical status, biochemical parameters, therapeutic regimens and outcome of Austrian LCHADD patients. Study design Clinical and outcome data including history, diagnosis, short- and long-term manifestations, growth, psychomotor development, hospitalizations, therapy of 14 Austrian patients with LCHADD were evaluated. Biochemically, we evaluated creatine kinase (CK) and acyl carnitine profiles. Results All LCHADD patients are homozygous for the common mutation. Three are siblings. Diagnosis was first established biochemically. Nine/14 (64%) were prematures, with IRDS occurring in six. In nine (64%), diagnosis was established through newborn screening, the remaining five (36%) were diagnosed clinically. Four pregnancies were complicated by HELLP syndrome, one by preeclampsia. In two, intrauterine growth retardation and placental insufficiency were reported. Five were diagnosed with hepatopathy at some point, seven with cardiomyopathy and eight with retinopathy, clinically relevant only in one patient. Polyneuropathy is only present in one. Three patients have a PEG, one is regularly fed via NG-tube. Growth is normal in all, as well as psychomotor development, except for two extremely premature girls. In 11 patients, 165 episodes with elevated creatine kinase concentrations were observed with 6-31 (median 14) per patient; three have shown no elevated CK concentrations. Median total carnitine on therapy was 19 μmol/l (range 11-61). For 14 patients, there have been 181 hospitalizations (median 9 per patient), comprising 1337 in-patient-days. All centres adhere to treatment with a fat-defined diet; patients have between 15% and 40% of their energy intake from fat (median 29%), out of which between 20% and 80% are medium-chain triglycerides (MCT) (median 62%). Four patients have been treated with heptanoate (C7). Conclusion Our data show LCHADD outcome can be favourable. Growth and psychomotor development is normal, except in two prematures. Frequency of CK measurements decreases with age, correlating with a decreasing number of hospitalizations. About 50% develop complications affecting different organ systems. There is no relevant difference between the patients treated in the respective centers. Concluding from single case reports, anaplerotic therapy with heptanoate should be further evaluated.
Collapse
Affiliation(s)
- Daniela Karall
- Medical University of Innsbruck, Clinic for Pediatrics, Inherited Metabolic Disorders, Anichstrasse 35, 6020, Innsbruck, Austria.
| | | | - Katharina Kogelnig
- Medical University of Innsbruck, Clinic for Pediatrics, Inherited Metabolic Disorders, Anichstrasse 35, 6020, Innsbruck, Austria.
| | | | - Esther M Maier
- Dr. von Hauner Children's Hospital, University of Munich, Munich, Germany.
| | | | | | | | | | - Sabine Scholl-Bürgi
- Medical University of Innsbruck, Clinic for Pediatrics, Inherited Metabolic Disorders, Anichstrasse 35, 6020, Innsbruck, Austria.
| |
Collapse
|
32
|
Karlstetter M, Scholz R, Rutar M, Wong WT, Provis JM, Langmann T. Retinal microglia: just bystander or target for therapy? Prog Retin Eye Res 2014; 45:30-57. [PMID: 25476242 DOI: 10.1016/j.preteyeres.2014.11.004] [Citation(s) in RCA: 394] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 11/20/2014] [Accepted: 11/25/2014] [Indexed: 10/24/2022]
Abstract
Resident microglial cells can be regarded as the immunological watchdogs of the brain and the retina. They are active sensors of their neuronal microenvironment and rapidly respond to various insults with a morphological and functional transformation into reactive phagocytes. There is strong evidence from animal models and in situ analyses of human tissue that microglial reactivity is a common hallmark of various retinal degenerative and inflammatory diseases. These include rare hereditary retinopathies such as retinitis pigmentosa and X-linked juvenile retinoschisis but also comprise more common multifactorial retinal diseases such as age-related macular degeneration, diabetic retinopathy, glaucoma, and uveitis as well as neurological disorders with ocular manifestation. In this review, we describe how microglial function is kept in balance under normal conditions by cross-talk with other retinal cells and summarize how microglia respond to different forms of retinal injury. In addition, we present the concept that microglia play a key role in local regulation of complement in the retina and specify aspects of microglial aging relevant for chronic inflammatory processes in the retina. We conclude that this resident immune cell of the retina cannot be simply regarded as bystander of disease but may instead be a potential therapeutic target to be modulated in the treatment of degenerative and inflammatory diseases of the retina.
Collapse
Affiliation(s)
- Marcus Karlstetter
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - Rebecca Scholz
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - Matt Rutar
- The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australian Capital Territory, Australia
| | - Wai T Wong
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jan M Provis
- The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australian Capital Territory, Australia
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany.
| |
Collapse
|
33
|
Hoffman DR, Hughbanks-Wheaton DK, Pearson NS, Fish GE, Spencer R, Takacs A, Klein M, Locke KG, Birch DG. Four-year placebo-controlled trial of docosahexaenoic acid in X-linked retinitis pigmentosa (DHAX trial): a randomized clinical trial. JAMA Ophthalmol 2014; 132:866-73. [PMID: 24805262 DOI: 10.1001/jamaophthalmol.2014.1634] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
IMPORTANCE X-linked retinitis pigmentosa is a severe inherited retinal degenerative disease with a frequency of 1 in 100,000 persons. Because no cure is available for this orphan disease and treatment options are limited, slowing of disease progression would be a meaningful outcome. OBJECTIVE To determine whether high-dose docosahexaenoic acid (DHA), an ω-3 polyunsaturated fatty acid, slows progression of X-linked retinitis pigmentosa measured by cone electroretinography (ERG). DESIGN, SETTING, AND PARTICIPANTS A 4-year, single-site, randomized, placebo-controlled, double-masked phase 2 clinical trial at a research center specializing in medical retina. Seventy-eight male patients diagnosed as having X-linked retinitis pigmentosa were randomized to DHA or placebo. Data were omitted for 2 patients with non-X-linked retinitis pigmentosa and 16 patients who were unable to follow protocol during the first year. The remaining participants were tested annually and composed a modified intent-to-treat cohort (DHA group, n = 33; placebo group, n = 27). INTERVENTIONS All participants received a multivitamin and were randomly assigned to oral DHA (30 mg/kg/d) or placebo. MAIN OUTCOMES AND MEASURES The primary outcome was the rate of loss of cone ERG function. Secondary outcomes were rod and maximal ERG amplitudes and cone ERG implicit times. Capsule counts and red blood cell DHA levels were assessed to monitor adherence. RESULTS Average (6-month to 4-year) red blood cell DHA levels were 4-fold higher in the DHA group than in the placebo group (P < .001). There was no difference between the DHA and placebo groups in the rate of cone ERG functional loss (0.028 vs 0.022 log µV/y, respectively; P = .30). No group differences were evident for change in rod ERG (P = .27), maximal ERG (P = .65), or cone implicit time (no change over 4 years). The rate of cone loss (ie, event rate) was markedly reduced compared with rates in previous studies. No severe treatment-emergent adverse events were found. CONCLUSIONS AND RELEVANCE Long-term DHA supplementation was not effective in slowing the loss of cone or rod ERG function associated with X-linked retinitis pigmentosa. Participant dropout and lower-than-expected disease event rate limited power to detect statistical significance. A larger sample size, longer trial, and attainment of a target blood DHA level (13%) would be desirable. While DHA supplementation at 30 mg/kg/d does not present serious adverse effects, routine monitoring of gastrointestinal tolerance is prudent. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00100230.
Collapse
Affiliation(s)
- Dennis R Hoffman
- Retina Foundation of the Southwest, Dallas, Texas2Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas
| | - Dianna K Hughbanks-Wheaton
- Retina Foundation of the Southwest, Dallas, Texas2Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas
| | - N Shirlene Pearson
- Pearson Statistical Consulting and Expert Witness Testimony, Richardson, Texas
| | | | | | | | - Martin Klein
- Retina Foundation of the Southwest, Dallas, Texas
| | | | - David G Birch
- Retina Foundation of the Southwest, Dallas, Texas2Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|
34
|
Karall D, Mair G, Albrecht U, Niedermayr K, Karall T, Schobersberger W, Scholl-Bürgi S. Sports in LCHAD Deficiency: Maximal Incremental and Endurance Exercise Tests in a 13-Year-Old Patient with Long-Chain 3-Hydroxy Acyl-CoA Dehydrogenase Deficiency (LCHADD) and Heptanoate Treatment. JIMD Rep 2014; 17:7-12. [PMID: 24997711 DOI: 10.1007/8904_2014_313] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 03/26/2014] [Accepted: 03/28/2014] [Indexed: 12/31/2022] Open
Abstract
Exercise and subsequent catabolism is a potential trigger for creatine kinase (CK) concentration increase (rhabdomyolysis) in patients with LCHADD, therefore we evaluated the clinical and biochemical stability under physical exertion conditions at the age of 13 years in a currently 14-year-old LCHADD patient treated with heptanoate.LCHADD was diagnosed during first decompensation at age 20 months. In the following 2 years, the patient had several episodes of rhabdomyolysis. Heptanoate 0.5-1 g/kg/day was started at 4 years, with no further CK elevations since. He is clinically stable, has retinopathy without vision impairment or polyneuropathy. Maximal incremental and endurance exercise tests were performed to evaluate both clinical and metabolic stability during and after exertion.Physical fitness was adequate for age (maximum blood lactate 7.0 mmol/L, appropriate lactate performance curve, maximum heart rate of 196 bpm, maximum power 139 Watt = 2.68 Watt/kg body weight). There were no signs of clinical (muscle pain, dark urine) or metabolic derangement (stable CK, acyl carnitine profiles, blood gas analyses) - neither after maximal incremental nor endurance exertion.This case illustrates that both under maximal incremental and endurance exertion, clinical and biochemical parameters remained stable in this currently 14-year-old LCHADD patient receiving heptanoate treatment.
Collapse
Affiliation(s)
- D Karall
- Division of Inherited Metabolic Disorders, Medical University Innsbruck, Clinic for Pediatrics I, Inherited Metabolic Disorders, Innsbruck, Austria,
| | | | | | | | | | | | | |
Collapse
|
35
|
Martin JM, Gillingham MB, Harding CO. Use of propofol for short duration procedures in children with long chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD) or trifunctional protein (TFP) deficiencies. Mol Genet Metab 2014; 112:139-42. [PMID: 24780638 PMCID: PMC4121654 DOI: 10.1016/j.ymgme.2014.03.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 03/26/2014] [Accepted: 03/26/2014] [Indexed: 11/28/2022]
Abstract
The medication propofol, commonly used for anesthesia, has been avoided in patients with mitochondrial fatty acid oxidation disorders (FAODs) due to concerns that it contains long-chain fatty acids (LCFAs), and because of reports of severe side effects in some critically ill patients receiving high-dose propofol infusions that mimic some of the symptoms regularly found in FAOD patients. In this secondary analysis, we examined the outcomes of 8 children with long-chain 3-hydroxyacyl CoA dehydrogenase (LCHAD) deficiency or trifunctional protein (TFP) deficiency who were repeatedly sedated for an electroretinogram (ERG) as part of a longitudinal study of the progression of chorioretinopathy commonly found in this population. A total of 39 sedated ERG procedures were completed using propofol for sedation. The propofol dosing, estimated total energy needs of the subject, and inpatient dietary intake recording were completed in 32 of these procedures. The LCFAs in the propofol provided approximately 1.0% of the average total daily energy needs. The sedation with propofol resulted in no adverse side effects and was safely used in this short duration procedure.
Collapse
Affiliation(s)
- Julie M Martin
- Department of Molecular & Medical Genetics, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Melanie B Gillingham
- Department of Molecular & Medical Genetics, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA; Graduate Programs in Human Nutrition, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Cary O Harding
- Department of Molecular & Medical Genetics, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| |
Collapse
|
36
|
Anderson S, Brooks SS. When the usual symptoms become an unusual diagnosis: a case report of trifunctional protein complex. Neonatal Netw 2013; 32:262-273. [PMID: 23835545 DOI: 10.1891/0730-0832.32.4.262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Disorders of mitochondrial fatty acid b-oxidation should be considered in any infant who presents with unexplained hypoglycemia and/or myopathy. Although disorders of trifunctional protein (TFP) complex including long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD) and mitochondrial TFP deficiencies are extremely rare, the combined incidence of mitochondrial fatty acid disorders is quite frequent. With the expansion of newborn screening, what were once considered uncommon disorders are being identified with increasing frequency in asymptomatic infants. The following case scenario presents an infant who developed symptoms prior to the completion of newborn screening. This fairly routine course for a late-preterm infant reveals an extremely rare inborn error of metabolism, LCHAD deficiency. An overview of TFP complex, the differential diagnoses as the case unfolds, diagnostic test results, acute care management, and short-term patient follow-up is presented. With experience, health care providers often become accustomed to and expect to see common things regularly. This case presents a scenario which, as it unfolds, appears to be quite common. It turns out, however, to be very uncommon.
Collapse
Affiliation(s)
- Sharon Anderson
- University of Medicine and Dentistry of New Jersey-School of Nursing, Newark, NJ 07101, USA.
| | | |
Collapse
|
37
|
Fletcher AL, Pennesi ME, Harding CO, Weleber RG, Gillingham MB. Observations regarding retinopathy in mitochondrial trifunctional protein deficiencies. Mol Genet Metab 2012; 106:18-24. [PMID: 22459206 PMCID: PMC3506186 DOI: 10.1016/j.ymgme.2012.02.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 02/23/2012] [Accepted: 02/23/2012] [Indexed: 12/31/2022]
Abstract
Although the retina is thought to primarily rely on glucose for fuel, inherited deficiency of one or more activities of mitochondrial trifunctional protein results in a pigmentary retinopathy leading to vision loss. Many other enzymatic deficiencies in fatty acid oxidation pathways have been described, none of which results in retinal complications. The etiology of retinopathy among patients with defects in trifunctional protein is unknown. Trifunctional protein is a heteroctomer; two genes encode the alpha and beta subunits of TFP respectively, HADHA and HADHB. A common mutation in HADHA, c.1528G>C, leads to a single amino acid substitution, p. Glu474Gln, and impairs primarily long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD) activity leading to LCHAD deficiency (LCHADD). Other mutations in HADHA or HADHB often lead to significant reduction in all three enzymatic activities and result in trifunctional protein deficiency (TFPD). Despite many similarities in clinical presentation and phenotype, there is growing evidence that they can result in different chronic complications. This review will outline the clinical similarities and differences between LCHADD and TFPD, describe the course of the associated retinopathy, propose a genotype/phenotype correlation with the severity of retinopathy, and discuss the current theories about the etiology of the retinopathy.
Collapse
Affiliation(s)
- Autumn L Fletcher
- Department of Molecular & Medical Genetics, School of Medicine, Oregon Health & Science University, Mail Code L-103, 3181 SW Sam Jackson Park Rd Portland, OR 97239, USA.
| | | | | | | | | |
Collapse
|
38
|
Okuläre Zeichen eines mitochondrialen trifunktionalen Proteindefekts. Ophthalmologe 2012; 109:277-82. [DOI: 10.1007/s00347-011-2480-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
39
|
Therapy for mitochondrial disorders: little proof, high research activity, some promise. Semin Fetal Neonatal Med 2011; 16:236-40. [PMID: 21676668 DOI: 10.1016/j.siny.2011.05.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mitochondrial disorders are a common group of metabolic diseases, the largest subgroup being the respiratory chain deficiencies. The most severe forms of mitochondrial dysfunction manifest in the neonatal period, rendering this group of patients the most challenging for therapy development. Heterogeneity of molecular backgrounds in the whole mitochondrial disease group has hindered therapy trials, but promising results are being gained from studies on animal models. Here I review strategies that have been tested or that can be proposed to be feasible as intervention. Many of these strategies aim to slow down the disease progression or are palliative in nature. However, currently very little evidence for any kind of therapeutic tools is available from double-blind controlled studies.
Collapse
|
40
|
Smith EC, El-Gharbawy A, Koeberl DD. Metabolic myopathies: clinical features and diagnostic approach. Rheum Dis Clin North Am 2011; 37:201-17, vi. [PMID: 21444020 DOI: 10.1016/j.rdc.2011.01.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The rheumatologist is frequently called on to evaluate patients with complaints of myalgia, muscle cramps, and fatigue. The evaluation of these patients presents a diagnostic challenge given the nonspecific and intermittent nature of their complaints, often leading to inappropriate diagnostic testing. When these symptoms are associated with physical exertion, a metabolic myopathy should be suspected Although inflammatory myopathies may present with similar features, such a pattern should prompt a thorough evaluation for an underlying metabolic myopathy. This review discusses the most common causes of metabolic myopathies and reviews the current diagnostic options available to the clinician.
Collapse
Affiliation(s)
- Edward C Smith
- Division of Pediatric Neurology, Department of Pediatrics, Duke University Medical Center, DUMC Box 3936, Durham, NC 27710, USA
| | | | | |
Collapse
|
41
|
Spiekerkoetter U. Mitochondrial fatty acid oxidation disorders: clinical presentation of long-chain fatty acid oxidation defects before and after newborn screening. J Inherit Metab Dis 2010; 33:527-32. [PMID: 20449660 DOI: 10.1007/s10545-010-9090-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2009] [Revised: 01/29/2010] [Accepted: 03/24/2010] [Indexed: 12/27/2022]
Abstract
The different long-chain fatty acid oxidation defects present with similar heterogeneous clinical phenotypes of different severity. Organs mainly affected comprise the heart, liver, and skeletal muscles. All symptoms are reversible with sufficient energy supply. In some long-chain fatty acid oxidation defects, disease-specific symptoms occur. Only in disorders of the mitochondrial trifunctional protein (TFP) complex, including long-chain 3-hydroxyacyl-coenzyme A (CoA) dehydrogenase (LCHAD) deficiency, neuropathy and retinopathy develop that are progressive and irreversible despite current treatment measures. In most long-chain fatty acid oxidation defects, no clear genotype-phenotype correlation exists due to molecular heterogeneity. However, some isolated mutations have been identified to be associated with only mild phenotypes, e.g., the V243A mutation in very-long-chain acyl-CoA dehydrogenase (VLCAD) deficiency. LCHAD deficiency is due to the prevalent homozygous 1528G>C mutation and presents with heterogeneous clinical phenotypes, suggesting the importance of other environmental and genetic factors. For some disorders, it was shown that residual enzyme activity measured in fibroblasts or lymphocytes correlated with severity of clinical phenotype. Implementation of newborn screening has significantly reduced morbidity and mortality of long-chain fatty acid oxidation defects. However, the severest forms of TFP deficiency are still highly associated with neonatal death. Newborn screening also identifies a great number of mildly affected patients who may never develop clinical symptoms throughout life. However, later-onset exercise-induced myopathic symptoms remain characteristic clinical features of long-chain fatty acid oxidation defects. Disease prevalence has increased with newborn screening.
Collapse
Affiliation(s)
- Ute Spiekerkoetter
- Department of General Pediatrics, University Children's Hospital, Duesseldorf, Germany.
| |
Collapse
|
42
|
Spiekerkoetter U, Bastin J, Gillingham M, Morris A, Wijburg F, Wilcken B. Current issues regarding treatment of mitochondrial fatty acid oxidation disorders. J Inherit Metab Dis 2010; 33:555-61. [PMID: 20830526 DOI: 10.1007/s10545-010-9188-1] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Revised: 07/27/2010] [Accepted: 08/06/2010] [Indexed: 12/13/2022]
Abstract
Treatment recommendations in mitochondrial fatty acid oxidation (FAO) defects are diverse. With implementation of newborn screening and identification of asymptomatic patients, it is necessary to define whom to treat and how strictly. We here discuss critical questions that are currently under debate. For some asymptomatic long-chain defects, long-chain fat restriction plays a minor role, and a normal diet may be introduced. For patients presenting only with myopathic symptoms, e.g., during exercise, treatment may be adapted to energy demand. As a consequence, patients with exercise-induced myopathy may be able to return to normal activity when provided with medium-chain triglycerides (MCT) prior to exercise. There is no need to limit participation in sports. Progression of retinopathy in disorders of the mitochondrial trifunctional protein complex is closely associated with hydroxyacylcarnitine accumulation. A strict low-fat diet with MCT supplementation is recommended to slow or prevent progression of chorioretinopathy. Additional docosahexanoic acid does not prevent the decline in retinal function but does promote nonspecific improvement in visual acuity and is recommended. There is no evidence that L-carnitine supplementation is beneficial. Thus, supplementation with L-carnitine in a newborn identified by screening with either a medium-chain or long-chain defect is not supported. With respect to the use of the odd-chain medium-chain triglyceride triheptanoin in myopathic phenotypes, randomized trials are needed to establish whether triheptanoin is more effective than even-chain MCT. With increasing pathophysiological knowledge, new treatment options have been identified and are being clinically evaluated. These include the use of bezafibrates in myopathic long-chain defects.
Collapse
Affiliation(s)
- Ute Spiekerkoetter
- Department of General Pediatrics, University Children's Hospital, Duesseldorf, Germany.
| | | | | | | | | | | |
Collapse
|
43
|
EFFECT OF FEEDING, EXERCISE AND GENOTYPE ON PLASMA 3-HYDROXYACYLCARNITINES IN CHILDREN WITH LCHAD DEFICIENCY. TOP CLIN NUTR 2009; 24:359-365. [PMID: 20589231 DOI: 10.1097/tin.0b013e3181c62182] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Chronic complications observed in patients with long-chain 3-hydroxyacylCoA dehydrogenase (LCHAD) or trifunctional protein (TFP) deficiency may be mediated by the accumulation of 3-hydroxy fatty acids or 3-hydroxyacylcarnitines. To understand variation in metabolite accumulation, their concentrations were measured by tandem mass spectrometry before and after a mixed meal and moderate intensity exercise. Subjects who were homozygous or heterozygous for the common mutation (c.1528G>C) in the TFP alpha subunit (LCHAD deficiency) had significantly higher 3-hydroxyacylcarnitines than subjects with TFP deficiency. Feeding a mixed meal significantly suppressed and exercise significantly increased plasma 3-hydroxyacylcarnitines concentrations.
Collapse
|
44
|
Spiekerkoetter U, Lindner M, Santer R, Grotzke M, Baumgartner MR, Boehles H, Das A, Haase C, Hennermann JB, Karall D, de Klerk H, Knerr I, Koch HG, Plecko B, Röschinger W, Schwab KO, Scheible D, Wijburg FA, Zschocke J, Mayatepek E, Wendel U. Treatment recommendations in long-chain fatty acid oxidation defects: consensus from a workshop. J Inherit Metab Dis 2009; 32:498-505. [PMID: 19452263 DOI: 10.1007/s10545-009-1126-8] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2008] [Revised: 03/24/2009] [Accepted: 03/25/2009] [Indexed: 12/13/2022]
Abstract
Published data on treatment of fatty acid oxidation defects are scarce. Treatment recommendations have been developed on the basis of observations in 75 patients with long-chain fatty acid oxidation defects from 18 metabolic centres in Central Europe. Recommendations are based on expert practice and are suggested to be the basis for further multicentre prospective studies and the development of approved treatment guidelines. Considering that disease complications and prognosis differ between different disorders of long-chain fatty acid oxidation and also depend on the severity of the underlying enzyme deficiency, treatment recommendations have to be disease-specific and depend on individual disease severity. Disorders of the mitochondrial trifunctional protein are associated with the most severe clinical picture and require a strict fat-reduced and fat-modified (medium-chain triglyceride-supplemented) diet. Many patients still suffer acute life-threatening events or long-term neuropathic symptoms despite adequate treatment, and newborn screening has not significantly changed the prognosis for these severe phenotypes. Very long-chain acyl-CoA dehydrogenase deficiency recognized in neonatal screening, in contrast, frequently has a less severe disease course and dietary restrictions in many patients may be loosened. On the basis of the collected data, recommendations are given with regard to the fat and carbohydrate content of the diet, the maximal length of fasting periods and the use of l-carnitine in long-chain fatty acid oxidation defects.
Collapse
Affiliation(s)
- U Spiekerkoetter
- Department of General Pediatrics, University Children's Hospital, Düsseldorf, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Spiekerkoetter U, Lindner M, Santer R, Grotzke M, Baumgartner MR, Boehles H, Das A, Haase C, Hennermann JB, Karall D, de Klerk H, Knerr I, Koch HG, Plecko B, Röschinger W, Schwab KO, Scheible D, Wijburg FA, Zschocke J, Mayatepek E, Wendel U. Management and outcome in 75 individuals with long-chain fatty acid oxidation defects: results from a workshop. J Inherit Metab Dis 2009; 32:488-97. [PMID: 19399638 DOI: 10.1007/s10545-009-1125-9] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2008] [Revised: 03/24/2009] [Accepted: 03/25/2009] [Indexed: 10/20/2022]
Abstract
At present, long-chain fatty acid oxidation (FAO) defects are diagnosed in a number of countries by newborn screening using tandem mass spectrometry. In the majority of cases, affected newborns are asymptomatic at time of diagnosis and acute clinical presentations can be avoided by early preventive measures. Because evidence-based studies on management of long-chain FAO defects are lacking, we carried out a retrospective analysis of 75 patients from 18 metabolic centres in Germany, Switzerland, Austria and the Netherlands with special regard to treatment and disease outcome. Dietary treatment is effective in many patients and can prevent acute metabolic derangements and prevent or reverse severe long-term complications such as cardiomyopathy. However, 38% of patients with very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency had intermittent muscle weakness and pain despite adhering to therapy. Seventy-six per cent of patients with disorders of the mitochondrial trifunctional protein (TFP)-complex including long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD) deficiency, had long-term myopathic symptoms. Of these, 21% had irreversible peripheral neuropathy and 43% had retinopathy. The main principle of treatment was a fat-reduced and fat-modified diet. Fat restriction differed among patients with different enzyme defects and was strictest in disorders of the TFP-complex. Patients with a medium-chain fat-based diet received supplementation of essential long-chain fatty acids. l-Carnitine was supplemented in about half of the patients, but in none of the patients with VLCAD deficiency identified by newborn screening. In summary, in this cohort the treatment regimen was adapted to the severity of the underlying enzyme defect and thus differed among the group of long-chain FAO defects.
Collapse
Affiliation(s)
- U Spiekerkoetter
- Department of General Pediatrics, University Children's Hospital, Düsseldorf, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Walter JH. Tolerance to fast: rational and practical evaluation in children with hypoketonaemia. J Inherit Metab Dis 2009; 32:214-7. [PMID: 19255872 DOI: 10.1007/s10545-009-1087-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2008] [Revised: 11/26/2008] [Accepted: 12/08/2008] [Indexed: 11/26/2022]
Abstract
Prolonged fasting in children with disorder of fat oxidation or ketone body synthesis can lead not only to hypoglycaemia but also to the accumulation of toxic metabolites. The length of time such patients can be safely fasted is important information for caregivers. Most children with MCAD deficiency when well can tolerate 'normal' periods without food, but in more severe disorders such as LCHAD deficiency even these may be associated with acute or chronic damage. Guidelines have been published for safe fasting periods in MCAD but not in other conditions. In the absence of such recommendations, a rational approach must be based on an understanding of the normal physiology of fasting in children of different ages and the pathophysiology associated with the child's particular disorder. Intercurrent infections pose a particular risk and may significantly reduce fasting tolerance.
Collapse
Affiliation(s)
- J H Walter
- Willink Biochemical Genetics Unit, Royal Manchester Children's Hospital, Hospital Road, Manchester, M27 4HA, UK.
| |
Collapse
|
47
|
Stopek D, Gitteau Lala E, Labarthe F, Le Lez ML, Majzoub S, Castelnau P, Pisella PJ. [Long-chain 3-hydroxyacyl CoA dehydrogenase deficiency and choroidal neovascularization]. J Fr Ophtalmol 2008; 31:993-8. [PMID: 19107076 DOI: 10.1016/s0181-5512(08)74746-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We report the case of a 9-year-old girl with a long-chain 3-hydroxyacyl CoA dehydrogenase (LCHAD) deficiency. This enzyme participates in mitochondrial fatty acid B-oxidation. Genetic fatty acid oxidation defects induce cellular energetic deficiency, and thus early life-threatening manifestations. An appropriate diet prevents these severe disorders. Nevertheless, LCHAD deficiency is the only B-oxidation enzymatic disorder that induces a chorioretinopathy, predominating at the posterior pole. We describe the first case of bilateral macular choroidal neovascularization. One eye presented a fibrovascular lesion. The other eye presented an active neovascularization stabilized by two dynamic phototherapies. The specificity of choroidal degeneration related to LCHAD deficiency remains unknown. Reviewing of literature and biochemical mechanisms suggests that fatty acid oxidative stress rather than a mitochondrial energetic defect is involved. For practical purposes, this report emphasizes the importance of ophthalmological follow-up of these patients.
Collapse
Affiliation(s)
- D Stopek
- Service d'Ophtalmologie, Hôpital Bretonneau, Tours.
| | | | | | | | | | | | | |
Collapse
|
48
|
Fahnehjelm KT, Holmström G, Ying L, Haglind CB, Nordenström A, Halldin M, Alm J, Nemeth A, von Döbeln U. Ocular characteristics in 10 children with long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency: a cross-sectional study with long-term follow-up. Acta Ophthalmol 2008; 86:329-37. [PMID: 18162058 DOI: 10.1111/j.1600-0420.2007.01121.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE To present long-term ocular complications and electroretinographic (ERG) findings in children with long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD) deficiency - a life-threatening metabolic disease - and the relation to age at diagnosis, treatment and other clinical parameters. METHODS Ten children with LCHAD deficiency underwent repeated ophthalmological evaluations including ERG. RESULTS All 10 children developed chorioretinal pathology. Regardless of age at diagnosis, initiation of treatment and age at examination, inter-individual differences were present. Profound chorioretinal atrophy, severe visual impairment and progressive myopia had developed in two teenagers. Milder chorioretinopathy with or without subnormal visual acuity was present in all other children. ERG was pathological in seven children. The chorioretinopathy often started in the peripapillary or perimacular areas. In one patient, unilateral visual impairment was associated with fibrosis. CONCLUSION Early diagnosis and adequate therapy might delay but not prevent the progression of retinal complications. Late diagnosis with severe symptoms at diagnosis, neonatal hypoglycaemia and frequent decompensations may increase the progression rate of the chorioretinopathy. LCHAD deficiency, a potentially lethal disease, is sometimes difficult to diagnose. Unusual chorioretinal findings should alert the ophthalmologist to the long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency, especially if there is a history of neonatal hypoglycaemia or failure to thrive.
Collapse
|
49
|
Gillingham MB, Purnell JQ, Jordan J, Stadler D, Haqq AM, Harding CO. Effects of higher dietary protein intake on energy balance and metabolic control in children with long-chain 3-hydroxy acyl-CoA dehydrogenase (LCHAD) or trifunctional protein (TFP) deficiency. Mol Genet Metab 2007; 90:64-9. [PMID: 16996288 PMCID: PMC2813195 DOI: 10.1016/j.ymgme.2006.08.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2006] [Revised: 08/07/2006] [Accepted: 08/07/2006] [Indexed: 11/19/2022]
Abstract
The incidence of overweight and obesity is increasing among children with long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD) or mitochondrial trifunctional (TFP) deficiency. Traditional treatment includes fasting avoidance and consumption of a low-fat, high-carbohydrate diet. A diet higher in protein and lower in carbohydrate may help to lower total energy intake while maintaining good metabolic control. To determine the short-term safety and efficacy of a high protein diet, subjects were admitted to the General Clinical Research Center and fed an ad-libitum high-protein diet and a high-carbohydrate diet for 6 days each using a randomized, crossover design. Nine subjects with LCHAD or TFP deficiency, age 7-14 were enrolled. Body composition was determined by DEXA. Total energy intake was evaluated daily. Resting energy expenditure and substrate utilization were determined by indirect calorimetry. Post-prandial metabolic responses of plasma glucose, insulin, leptin, ghrelin, acylcarnitines, and triglyceride were determined in response to a liquid meal. Subjects had a higher fat mass, lower lean mass and higher plasma leptin levels compared to reference values. While on the high protein diet energy consumption was an average of 50 kcals/day lower (p = 0.02) and resting energy expenditure was an average of 170 kcals/day higher (p = 0.05) compared to the high carbohydrate diet. Short-term higher protein diets were safe, well tolerated, and resulted in lowered energy intake and increased energy expenditure than the standard high-carbohydrate diet. Long-term studies are needed to determine whether higher protein diets will reduce the risk of overweight and obesity in children with LCHAD or TFP deficiency.
Collapse
Affiliation(s)
- Melanie B Gillingham
- The Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Metabolic control during exercise with and without medium-chain triglycerides (MCT) in children with long-chain 3-hydroxy acyl-CoA dehydrogenase (LCHAD) or trifunctional protein (TFP) deficiency. Mol Genet Metab 2006; 89:58-63. [PMID: 16876451 DOI: 10.1016/j.ymgme.2006.06.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2006] [Revised: 06/09/2006] [Accepted: 06/09/2006] [Indexed: 11/17/2022]
Abstract
Exercise induced rhabdomyolysis is a complication of long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD) and mitochondrial trifunctional protein (TFP) deficiency that frequently leads to exercise avoidance. Dietary therapy for most subjects includes medium-chain triglyceride (MCT) supplementation but analysis of diet records indicates that the majority of patients consume oral MCT only with breakfast and at bedtime. We hypothesized that MCT immediately prior to exercise would provide an alternative fuel source during that bout of exercise and improve exercise tolerance in children with LCHAD deficiency. Nine subjects completed two 45 min moderate intensity (60-70% predicted maximum heart rate (HR)) treadmill exercise tests. Subjects were given 4 oz of orange juice alone or orange juice and 0.5 g MCT per kg lean body mass, 20 min prior to exercise in a randomized cross-over design. ECG and respiratory gas exchange including respiratory quotient (RQ) were monitored. Blood levels of acylcarnitines, creatine kinase, lactate, and beta-hydroxybutyrate were measured prior to and immediately after exercise, and again following 20 min rest. Creatine kinase and lactate levels did not change with exercise. There was no significant difference in RQ between the two exercise tests but there was a decrease in steady-state HR following MCT supplementation. Cumulative long-chain 3-hydroxyacylcarnitines were 30% lower and beta-hydroxybutyrate was three-fold higher after the MCT-pretreated exercise test compared to the test with orange juice alone. Coordinating MCT supplementation with periods of increased activity may improve the metabolic control of children with LCHAD and TFP deficiency following exercise.
Collapse
|