1
|
Daly A, Evans S, Pinto A, Ashmore C, MacDonald A. A 12-month, longitudinal, intervention study examining a tablet protein substitute preparation in the management of tyrosinemia. Mol Genet Metab Rep 2024; 40:101119. [PMID: 39081551 PMCID: PMC11286985 DOI: 10.1016/j.ymgmr.2024.101119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 08/02/2024] Open
Abstract
Protein substitutes (PS) without tyrosine (Tyr) and phenylalanine (Phe), are an essential source of synthetic protein in the treatment of tyrosinemia (HT). In the UK, the only available protein substitutes for HT are Tyr/ Phe free amino acid liquid or powders or formulations based on glycomacropeptide (CGMP). A tablet Tyr/ Phe free amino acid supplement (AAT) has now been introduced. The aim of this two-part prospective, longitudinal intervention study was to assess the efficacy, acceptability, and tolerance of AAT in children aged >8 years with HTI. Part 1: was a 28-day acceptability/ tolerance study, part 2, was a 12-month extension study examining efficacy of AAT. Anthropometry and blood Tyr/ Phe were assessed. All subjects were taking NTBC [2-(2-nitro-4-triflourothybenzoyl) cyclohexane-1, 3-dione] with a Tyr restricted diet. Eight subjects with HTI were recruited 4 boys, and 4 girls with a median age of 14.3y (range 10.4-17.3); 3 were Caucasian and 5 of Pakistani origin. The median (range) protein equivalent from PS was 60 g/d (50-60), natural protein 20 g/d (15-30), and NTBC 30 mg/d (25-80). No subjects were taking Phe supplements. Five (63%) subjects completed part 1, with 4 taking all their PS requirements as AAT. Subjects reported AAT were tasteless and had no odour. No adverse gastrointestinal symptoms were recorded, with two reporting improvements in abdominal discomfort. At 12 months, 4 subjects had a non-significant decrease in blood Tyr/ Phe compared to the 12 months pre-treatment. Median blood Tyr (μmol/ L) pre-intervention was 500 (320-590); and at 12 months, 450 (290-530). Median blood Phe (μmol/L) pre-intervention was 40 (30-40); and at 12 months 30 (30-50). Median height z scores remained unchanged, but there was a small decrease in weight z score (pre-study weight - 0.1 (-1.4 to1.1), 12 m - 0.3 (-1.4 to 1.3) and BMI (pre- study BMI 0.2 (-2 to 1.4), and 12 m, -0.1 (-2.5 to 1.5)). Conclusion AAT were useful for some adolescents with HTI who struggled with the taste and volume of conventional powdered and liquid PS.
Collapse
Affiliation(s)
- Anne Daly
- Birmingham Women 's and Children's Hospital, Steelhouse Lane, Birmingham, UK
| | - Sharon Evans
- Birmingham Women 's and Children's Hospital, Steelhouse Lane, Birmingham, UK
| | - Alex Pinto
- Birmingham Women 's and Children's Hospital, Steelhouse Lane, Birmingham, UK
| | - Catherine Ashmore
- Birmingham Women 's and Children's Hospital, Steelhouse Lane, Birmingham, UK
| | - Anita MacDonald
- Birmingham Women 's and Children's Hospital, Steelhouse Lane, Birmingham, UK
| |
Collapse
|
2
|
Kehar M, Sen Sarma M, Seetharaman J, Jimenez Rivera C, Chakraborty P. Decoding hepatorenal tyrosinemia type 1: Unraveling the impact of early detection, NTBC, and the role of liver transplantation. CANADIAN LIVER JOURNAL 2024; 7:54-63. [PMID: 38505790 PMCID: PMC10946188 DOI: 10.3138/canlivj-2023-0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/23/2023] [Indexed: 03/21/2024]
Abstract
Hepatorenal tyrosinemia type 1 (HT-1) is a rare autosomal recessive disease that results from a deficiency of fumaryl acetoacetate hydrolase (FAH), a critical enzyme in the catabolic pathway for tyrosine. This leads to the accumulation of toxic metabolites such as fumaryl and maleylacetoacetate, which can damage the liver, kidneys, and nervous system. The discovery of 2-[2-nitro-4-trifluoromethylbenzoyl]-1,3-cyclohexanedione (NTBC or nitisinone) has significantly improved the management of HT-1, particularly when initiated before the onset of symptoms. Therefore, newborn screening for HT-1 is essential for timely diagnosis and prompt treatment. The analysis of succinyl acetone (SA) in dried blood spots of newborns followed by quantification of SA in blood or urine for high-risk neonates has excellent sensitivity and specificity for the diagnosis of HT-1. NTBC combined with dietary therapy, if initiated early, can provide liver transplant (LT) free survival and reduce the risk of hepatocellular carcinoma (HCC). Patients failing medical treatment (eg, due to non-adherence), and who develop acute liver failure (ALF), have HCC or evidence of histologically proven dysplastic liver nodule(s), or experience poor quality of life secondary to severe dietary restrictions are currently indicated for LT. Children with HT-1 require frequent monitoring of liver and renal function to assess disease progression and treatment compliance. They are also at risk of long-term neurocognitive impairment, which highlights the need for neurocognitive assessment and therapy.
Collapse
Affiliation(s)
- Mohit Kehar
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Children Hospital of Eastern Ontario, Ottawa, Canada
| | - Moinak Sen Sarma
- Department of Pediatric Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Jayendra Seetharaman
- Division of Pediatric Gastroenterology and Hepatology, Christian Medical College, Vellore, India
| | - Carolina Jimenez Rivera
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Children Hospital of Eastern Ontario, Ottawa, Canada
| | - Pranesh Chakraborty
- Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa, Ottawa, Ontario, Canada
- Newborn Screening Ontario, Ottawa, Ontario, Canada
| |
Collapse
|
3
|
Szigetvari PD, Patil S, Birkeland E, Kleppe R, Haavik J. The effects of phenylalanine and tyrosine levels on dopamine production in rat PC12 cells. Implications for treatment of phenylketonuria, tyrosinemia type 1 and comorbid neurodevelopmental disorders. Neurochem Int 2023; 171:105629. [PMID: 37865339 DOI: 10.1016/j.neuint.2023.105629] [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: 08/21/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/23/2023]
Abstract
Phenylketonuria (PKU) is an autosomal recessive metabolic disorder caused by mutations in the phenylalanine hydroxylase (PAH) gene, resulting in phenylalanine accumulation and impaired tyrosine production. In Tyrosinemia type 1 (TYRSN1) mutations affect fumarylacetoacetate hydrolase, leading to accumulation of toxic intermediates of tyrosine catabolism. Treatment of TYRSN1 with nitisinone results in extreme tissue levels of tyrosine. Although PKU and TYRSN1 have opposite effects on tyrosine levels, both conditions have been associated with neuro-psychiatric symptoms typically present in ADHD, possibly indicating an impaired dopamine (DA) synthesis. However, concrete in vivo data on the possible molecular basis for disrupted DA production under disease mimicking conditions have been lacking. In pursuit to uncover associated molecular mechanisms, we exposed an established, DA producing cell line (PC12) to different concentrations of phenylalanine and tyrosine in culture media. We measured the effects on viability, proteomic composition, tyrosine, DA and tyrosine hydroxylase (TH) levels and TH phosphorylation. TH catalyzes the rate-limiting step in DA synthesis. High extracellular levels of phenylalanine depleted cells of intracellular tyrosine and DA. Compared to physiological levels (75 μM), either low (35 μM) or high concentrations of tyrosine (275 or 835 μM) decreased cellular DA, TH protein, and its phosphorylation levels. Using deep proteomic analysis, we identified multiple proteins, biological processes and pathways that were altered, including enzymes and transporters involved in amino acid metabolism. Using this information and published data, we developed a mathematical model to predict how extracellular levels of aromatic amino acids can affect the cellular synthesis of DA via different mechanisms. Together, these data provide new information about the normal regulation of neurotransmitter synthesis and how this may be altered in neurometabolic disorders, such as PKU and TYRSN1, with implications for the treatment of cognitive symptoms resulting from comorbid neurodevelopmental disorders.
Collapse
Affiliation(s)
| | - Sudarshan Patil
- Department of Biomedicine, University of Bergen, 5009, Bergen, Norway.
| | - Even Birkeland
- Department of Genetic Research & Bioinformatics, Norwegian Institute of Public Health, Bergen, Norway; The Proteomics Facility of the University of Bergen (PROBE), University of Bergen, Bergen, Norway
| | - Rune Kleppe
- Norwegian Centre for Maritime- and Diving Medicine, Department of Occupational Medicine, Haukeland University Hospital, 5021, Bergen, Norway.
| | - Jan Haavik
- Department of Biomedicine, University of Bergen, 5009, Bergen, Norway; Bergen Center of Brain Plasticity, Division of Psychiatry, Haukeland University Hospital, Norway.
| |
Collapse
|
4
|
Barone H, Elgen IB, Bliksrud YT, Vangsøy Hansen E, Skavhellen RR, Furevik MI, Haavik J. Case report: ADHD and prognosis in tyrosinemia type 1. Front Psychiatry 2023; 14:1213590. [PMID: 37533886 PMCID: PMC10392124 DOI: 10.3389/fpsyt.2023.1213590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/26/2023] [Indexed: 08/04/2023] Open
Abstract
Neurometabolic disorders such as tyrosinemia type 1 (TYRSN1) may interfere with brain metabolism and show symptoms of attention-deficit hyperactivity disorder (ADHD) in patients treated with the enzyme inhibitor nitisinone [2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione, NTBC]. It has been reported that ADHD treatment improves treatment compliance, which is imperative for the long-term prognosis of patients with TYRSN1. In this study, we report the case of a male patient who was diagnosed with TYRSN1 at 3 months of age and was subsequently treated with NTBC, restricted protein intake, and amino acids supplementation. At 7 years of age, he was referred for neuropsychiatric assessment, diagnosed with ADHD, and treated with methylphenidate. The effects of the treatment were monitored via parental interviews, questionnaires covering ADHD symptoms, and a continuous performance test. A reduction in ADHD symptoms, particularly inattentiveness, was observed across all measures. The early identification of ADHD and the treatment of neurometabolic disorders, such as TYRSN1, may be important from a lifetime perspective as this may improve the prognosis of the medical condition as well.
Collapse
Affiliation(s)
- Helene Barone
- Regional Resource Center for Autism, ADHD and Tourette Syndrome, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Irene Bircow Elgen
- Department of Child and Adolescent Psychiatry, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | | | | | - Rita Rigmor Skavhellen
- Department of Child and Adolescent Psychiatry, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Magne Ivar Furevik
- Department of Child and Adolescent Psychiatry, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Jan Haavik
- Bergen Center of Brain Plasticity, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
- Department of Biomedicine, University of Bergen, Bergen, Norway
| |
Collapse
|
5
|
Davison AS, Norman BP. Alkaptonuria – Past, present and future. Adv Clin Chem 2023. [DOI: 10.1016/bs.acc.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
6
|
Daly A, Adam S, Allen H, Ash J, Dale C, Dixon M, Dunlop C, Ellerton C, Evans S, Firman S, Ford S, Freedman F, Gribben J, Howe S, Khan F, McDonald J, McStravick N, Nguyen P, Oxley N, Skeath R, Simpson E, Terry A, Woodall A, White L, MacDonald A. UK Dietary Practices for Tyrosinaemias: Time for Change. Nutrients 2022; 14:nu14245202. [PMID: 36558364 PMCID: PMC9787818 DOI: 10.3390/nu14245202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022] Open
Abstract
In the UK, different dietary systems are used to calculate protein or tyrosine/phenylalanine intake in the dietary management of hereditary tyrosinaemia, HTI, II and III (HT), with no systematic evidence comparing the merits and inadequacies of each. This study aimed to examine the current UK dietary practices in all HTs and, using Delphi methodology, to reach consensus agreement about the best dietary management system. Over 12 months, five meetings were held with UK paediatric and adult dietitians working in inherited metabolic disorders (IMDs) managing HTs. Eleven statements on the dietary system for calculating protein or tyrosine/phenylalanine intake were discussed. Dietitians from 12 of 14 IMD centres caring for HT patients participated, and 7/11 statements were agreed with one Delphi round. Nine centres (three abstentions) supported a 1 g protein exchange system for all foods except fruit and vegetables. The same definitions used in the UK for phenylketonuria (PKU) were adopted to define when to calculate foods as part of a protein exchange system or permit them without measurement. Fruit and vegetables contain a lower amount of tyrosine/phenylalanine per 1 g of protein than animal and cereal foods. The correlation of tyrosine vs. phenylalanine (mg/100 g) for vegetables and fruits was high (r = 0.9). In Delphi round 2, agreement was reached to use the tyrosine/phenylalanine analyses of fruits/vegetables, for their allocation within the HT diet. This allowed larger portion sizes of measured fruits and vegetables and increased the variety of fruit and vegetables that could be eaten without measurement. In HTs, a combined dietary management system will be used: 1 g protein exchanges for cereal and milk protein sources and tyrosine/phenylalanine exchanges for fruit and vegetables. Intensive, systematic communication with IMD dietitians and reappraisal of the evidence has redefined and harmonised HT dietary practice across the UK.
Collapse
Affiliation(s)
- Anne Daly
- Birmingham Women’s and Children’s Hospital, NHS Foundation Trust, Steelhouse Lane, Birmingham B4 6NH, UK
- Correspondence:
| | - Sarah Adam
- Royal Hospital for Children, Glasgow G51 4TF, UK
| | - Heather Allen
- Sheffield Children’s NHS Foundation Trust, Sheffield S10 2TH, UK
| | - Jane Ash
- University Hospital of Wales, Cardiff CF4 4XW, UK
| | - Clare Dale
- University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2TH, UK
| | - Marjorie Dixon
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | | | - Charlotte Ellerton
- University College London Hospitals NHS Foundation Trust, London WC1N 3BG, UK
| | - Sharon Evans
- Birmingham Women’s and Children’s Hospital, NHS Foundation Trust, Steelhouse Lane, Birmingham B4 6NH, UK
| | - Sarah Firman
- Guy’s and St Thomas’ NHS Foundation Trust, London SE1 7EU, UK
| | - Suzanne Ford
- Southmead Hospital North Bristol Trust, Bristol BS10 5NB, UK
| | - Francine Freedman
- University College London Hospitals NHS Foundation Trust, London WC1N 3BG, UK
| | - Joanna Gribben
- Evelina London Children’s Healthcare, London SE1 7EH, UK
| | - Sara Howe
- University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2TH, UK
| | - Farzana Khan
- Bradford Teaching Hospitals, NHS Foundation Trust, Bradford BD5 0NA, UK
| | - Joy McDonald
- Belfast Health and Social Care Trust, Belfast BT9 7AB, UK
| | | | - Patty Nguyen
- University College London Hospitals NHS Foundation Trust, London WC1N 3BG, UK
| | - Natalia Oxley
- Bradford Teaching Hospitals, NHS Foundation Trust, Bradford BD5 0NA, UK
| | - Rachel Skeath
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Emma Simpson
- Royal Manchester Children’s Hospital, Manchester M13 9WL, UK
| | - Allyson Terry
- Alder Hey Children’s NHS Foundation Trust, Liverpool L12 2AP, UK
| | - Alison Woodall
- Salford Royal NHS Foundation Trust, Manchester M6 8HD, UK
| | - Lucy White
- Sheffield Children’s NHS Foundation Trust, Sheffield S10 2TH, UK
| | - Anita MacDonald
- Birmingham Women’s and Children’s Hospital, NHS Foundation Trust, Steelhouse Lane, Birmingham B4 6NH, UK
| |
Collapse
|
7
|
Bhushan S, Noble C, Balouch F, Lewindon P, Lampe G, Hodgkinson P, McGill J, Ee L. Hepatocellular carcinoma requiring liver transplantation in hereditary tyrosinemia type 1 despite nitisinone therapy and α1-fetoprotein normalization. Pediatr Transplant 2022; 26:e14334. [PMID: 35698261 DOI: 10.1111/petr.14334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 05/23/2022] [Accepted: 05/26/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Hereditary tyrosinemia type 1 is a rare metabolic condition associated with an increased risk of hepatocellular carcinoma. Nitisinone (2-[2-nitro-4-trifluoromethylbenzoyl]-1,3-cyclohexanedione, NTBC) treatment has reduced but not eliminated the risk. The delayed initiation of nitisinone treatment, and persistently abnormal α1-fetoprotein (AFP) levels are recognized to be risk factors for late-onset hepatocellular carcinoma. We report three children diagnosed and treated with nitisinone since infancy who developed hepatocellular carcinoma despite long-term normalization of AFP. METHODS A retrospective review of all patients with tyrosinemia on nitisinone managed at our center was undertaken. Patient demographics, age at diagnosis, duration of therapy, timing of AFP normalization, and radiographic imaging findings were noted. RESULTS Three patients at our center with tyrosinemia type 1 developed hepatocellular carcinoma 9-13 years after diagnosis despite long-term nitisinone therapy and normalization of AFP. Two patients developed new nodules on imaging with an elevation of AFP leading to the diagnosis and subsequent liver transplant. The third patient proceeded with liver transplant because of a very nodular liver and increasing splenomegaly despite normal AFP and no change in surveillance gadoxetate magnetic resonance imaging. Early hepatocellular carcinoma was found in her liver explant. All three patients were cirrhotic at diagnosis. CONCLUSIONS Patients with hereditary tyrosinemia type 1, especially those already cirrhotic at diagnosis, remain at high risk of developing hepatocellular carcinoma despite long-term nitisinone therapy and AFP normalization, and warrant close monitoring and surveillance.
Collapse
Affiliation(s)
- Shreya Bhushan
- Department of Gastroenterology, Hepatology and Liver Transplantation, Queensland Children's Hospital, Brisbane, Queensland, Australia
| | - Charlton Noble
- Department of Gastroenterology, Hepatology and Liver Transplantation, Queensland Children's Hospital, Brisbane, Queensland, Australia
| | - Fariha Balouch
- Department of Gastroenterology, Hepatology and Liver Transplantation, Queensland Children's Hospital, Brisbane, Queensland, Australia
| | - Peter Lewindon
- Department of Gastroenterology, Hepatology and Liver Transplantation, Queensland Children's Hospital, Brisbane, Queensland, Australia
| | - Guy Lampe
- Department of Anatomical Pathology, Central Laboratory Pathology Queensland, Herston, Queensland, Australia
| | - Peter Hodgkinson
- Queensland Liver Transplant Service, Princess Alexandra Hospital and Queensland Children's Hospital, Brisbane, Queensland, Australia
| | - Jim McGill
- Department of Chemical Pathology, Central Laboratory Pathology Queensland, Herston, Queensland, Australia
| | - Looi Ee
- Department of Gastroenterology, Hepatology and Liver Transplantation, Queensland Children's Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
8
|
van Vliet K, van Ginkel WG, Jahja R, Daly A, MacDonald A, Santra S, De Laet C, Goyens PJ, Vara R, Rahman Y, Cassiman D, Eyskens F, Timmer C, Mumford N, Gissen P, Bierau J, van Hasselt PM, Wilcox G, Morris AAM, Jameson EA, de la Parra A, Arias C, Garcia MI, Cornejo V, Bosch AM, Hollak CEM, Rubio‐Gozalbo ME, Brouwers MCGJ, Hofstede FC, de Vries MC, Janssen MCH, van der Ploeg AT, Langendonk JG, Huijbregts SCJ, van Spronsen FJ. Neurocognitive outcome and mental health in children with tyrosinemia type 1 and phenylketonuria: A comparison between two genetic disorders affecting the same metabolic pathway. J Inherit Metab Dis 2022; 45:952-962. [PMID: 35722880 PMCID: PMC9540223 DOI: 10.1002/jimd.12528] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/23/2022] [Accepted: 06/15/2022] [Indexed: 12/04/2022]
Abstract
Tyrosinemia type 1 (TT1) and phenylketonuria (PKU) are both inborn errors of phenylalanine-tyrosine metabolism. Neurocognitive and behavioral outcomes have always featured in PKU research but received less attention in TT1 research. This study aimed to investigate and compare neurocognitive, behavioral, and social outcomes of treated TT1 and PKU patients. We included 33 TT1 patients (mean age 11.24 years; 16 male), 31 PKU patients (mean age 10.84; 14 male), and 58 age- and gender-matched healthy controls (mean age 10.82 years; 29 male). IQ (Wechsler-subtests), executive functioning (the Behavioral Rating Inventory of Executive Functioning), mental health (the Achenbach-scales), and social functioning (the Social Skills Rating System) were assessed. Results of TT1 patients, PKU patients, and healthy controls were compared using Kruskal-Wallis tests with post-hoc Mann-Whitney U tests. TT1 patients showed a lower IQ and poorer executive functioning, mental health, and social functioning compared to healthy controls and PKU patients. PKU patients did not differ from healthy controls regarding these outcome measures. Relatively poor outcomes for TT1 patients were particularly evident for verbal IQ, BRIEF dimensions "working memory", "plan and organize" and "monitor", ASEBA dimensions "social problems" and "attention problems", and for the SSRS "assertiveness" scale (all p values <0.001). To conclude, TT1 patients showed cognitive impairments on all domains studied, and appeared to be significantly more affected than PKU patients. More attention should be paid to investigating and monitoring neurocognitive outcome in TT1 and research should focus on explaining the underlying pathophysiological mechanism.
Collapse
Affiliation(s)
- Kimber van Vliet
- Division of Metabolic DiseasesUniversity of Groningen, University Medical Center Groningen, Beatrix Children's HospitalGroningenThe Netherlands
| | - Willem G. van Ginkel
- Division of Metabolic DiseasesUniversity of Groningen, University Medical Center Groningen, Beatrix Children's HospitalGroningenThe Netherlands
| | - Rianne Jahja
- Division of Metabolic DiseasesUniversity of Groningen, University Medical Center Groningen, Beatrix Children's HospitalGroningenThe Netherlands
| | - Anne Daly
- Birmingham Children's HospitalBirminghamUK
| | | | | | - Corinne De Laet
- Hôpital Universitaire des Enfants Reine FabiolaUniversité Libre de BruxellesBrusselsBelgium
| | - Philippe J. Goyens
- Hôpital Universitaire des Enfants Reine FabiolaUniversité Libre de BruxellesBrusselsBelgium
| | | | | | - David Cassiman
- University Hospital Gasthuisberg, University of LeuvenLeuvenBelgium
| | - Francois Eyskens
- Kon. Mathilde Moeder‐ en KindcentrumUniversity Hospital of AntwerpAntwerpBelgium
| | | | - Nicky Mumford
- NIHR Great Ormond Street Hospital Biomedical Research CentreUniversity College LondonLondonUK
| | - Paul Gissen
- NIHR Great Ormond Street Hospital Biomedical Research CentreUniversity College LondonLondonUK
| | - Jörgen Bierau
- Maastricht University Medical CenterMaastrichtThe Netherlands
| | - Peter M. van Hasselt
- Wilhelmina Children's HospitalUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Gisela Wilcox
- School of Medical Sciences, Faculty of Biology Medicine & HealthUniversity of ManchesterManchesterUK
- The Mark Holland Metabolic Unit, Salford Royal Foundation NHS TrustSalfordUK
| | - Andrew A. M. Morris
- Willink Metabolic Unit, Manchester Centre for Genomic MedicineManchester University Hospitals NHS Foundation Trust, St Mary's HospitalManchesterUK
| | - Elisabeth A. Jameson
- Willink Metabolic Unit, Manchester Centre for Genomic MedicineManchester University Hospitals NHS Foundation Trust, St Mary's HospitalManchesterUK
| | - Alicia de la Parra
- Laboratory of Genetics and Metabolic Disease (LABGEM), Institute of Nutrition and Food Technology (INTA)University of ChileSantiagoChile
| | - Carolina Arias
- Laboratory of Genetics and Metabolic Disease (LABGEM), Institute of Nutrition and Food Technology (INTA)University of ChileSantiagoChile
| | - Maria I. Garcia
- Laboratory of Genetics and Metabolic Disease (LABGEM), Institute of Nutrition and Food Technology (INTA)University of ChileSantiagoChile
| | - Veronica Cornejo
- Laboratory of Genetics and Metabolic Disease (LABGEM), Institute of Nutrition and Food Technology (INTA)University of ChileSantiagoChile
| | - Annet M. Bosch
- Department of Pediatrics, Division of Metabolic Disorders, Emma Children's Hospital, Amsterdam Gastroenterology, Endocrinology & Metabolism, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Carla E. M. Hollak
- Department of Internal MedicineDivision of Endocrinology and Metabolism, Amsterdam UMC ‐ Location AMCAmsterdamThe Netherlands
| | - M. Estela Rubio‐Gozalbo
- Departments of Pediatrics and Laboratory Genetic Metabolic DiseasesMaastricht University Medical HospitalMaastrichtThe Netherlands
| | - Martijn C. G. J. Brouwers
- Department of Internal Medicine, Division of Endocrinology and Metabolic DiseaseMaastricht University Medical CentreMaastrichtThe Netherlands
- CARIM School for Cardiovascular DiseasesMaastricht UniversityMaastrichtThe Netherlands
| | - Floris C. Hofstede
- Wilhelmina Children's HospitalUniversity Medical Center UtrechtUtrechtThe Netherlands
| | | | | | - Ans T. van der Ploeg
- Departments of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Janneke G. Langendonk
- Department of Internal medicine, Center for Lysosomal and Metabolic Diseases, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Stephan C. J. Huijbregts
- University of Leiden, Clinical Child and Adolescent Studies: Neurodevelopmental DisordersLeidenThe Netherlands
| | - Francjan J. van Spronsen
- Division of Metabolic DiseasesUniversity of Groningen, University Medical Center Groningen, Beatrix Children's HospitalGroningenThe Netherlands
| |
Collapse
|
9
|
Kawabata K, Kido J, Yoshida T, Matsumoto S, Nakamura K. A case report of two siblings with hypertyrosinemia type 1 presenting with hepatic disease with different onset time and severity. Mol Genet Metab Rep 2022; 32:100892. [PMID: 35800472 PMCID: PMC9254452 DOI: 10.1016/j.ymgmr.2022.100892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/25/2022] [Accepted: 06/25/2022] [Indexed: 12/02/2022] Open
Abstract
Hereditary tyrosinemia type 1 (HT1) is an autosomal recessive disorder caused by a defect in fumarylacetoacetate hydroxylase (FAH) encoded by the FAH gene. Patients with HT1 disorder present with increased blood tyrosine, succinyl acetoacetate, and succinyl acetone levels, and develop clinical manifestations including liver failure, kidney tubular dysfunction, growth failure, rickets, pseudo-porphyric crises, and hepatocellular carcinoma. We encountered two siblings with HT1. Among the siblings, the elder brother developed acute liver failure with coagulopathy at the age of 2 months and was rescued by liver transplantation (LT) following combination therapy with continuous hemodiafiltration and plasma exchange. The younger sister was followed up from the prenatal period for signs of HT1 due to prior history of the condition in her sibling. She was initially considered a carrier of HT1 owing to the lack of overt signs of the disease and negative urine screening for succinyl acetone (SA). She was eventually diagnosed with HT1 because of liver disorder at 9 months of age, associated with a positive urine SA result. Her disease state was controlled by treatment with nitisinone (NTBC). DNA analysis of both siblings identified heterozygous status for a previously reported FAH pathogenic allele (c.782C > T) and a novel likely pathogenic variant (c.688C.G). The siblings have stable lives with no developmental delay or impaired growth. NTBC treatment is effective in preventing the progression of liver and kidney diseases. However, even in cases treated without LT, clinicians should follow up the clinical outcomes over long term, as patients may require LT when developing complications, such as hepatocellular carcinoma.
Collapse
Affiliation(s)
- Kazuo Kawabata
- Department of Pediatrics, Kumamoto University Hospital, Kumamoto, Japan
| | - Jun Kido
- Department of Pediatrics, Kumamoto University Hospital, Kumamoto, Japan
- Department of Pediatrics, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
- Corresponding author at: Department of Pediatrics, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto City, Kumamoto 860-8556, Japan.
| | - Takanobu Yoshida
- Department of Pediatrics, Kumamoto University Hospital, Kumamoto, Japan
- Department of Pediatrics, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Shirou Matsumoto
- Department of Pediatrics, Kumamoto University Hospital, Kumamoto, Japan
- Department of Pediatrics, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kimitoshi Nakamura
- Department of Pediatrics, Kumamoto University Hospital, Kumamoto, Japan
- Department of Pediatrics, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
10
|
Nicolas CT, VanLith CJ, Hickey RD, Du Z, Hillin LG, Guthman RM, Cao WJ, Haugo B, Lillegard A, Roy D, Bhagwate A, O'Brien D, Kocher JP, Kaiser RA, Russell SJ, Lillegard JB. In vivo lentiviral vector gene therapy to cure hereditary tyrosinemia type 1 and prevent development of precancerous and cancerous lesions. Nat Commun 2022; 13:5012. [PMID: 36008405 PMCID: PMC9411607 DOI: 10.1038/s41467-022-32576-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/08/2022] [Indexed: 11/23/2022] Open
Abstract
Conventional therapy for hereditary tyrosinemia type-1 (HT1) with 2-(2-nitro-4-trifluoromethylbenzoyl)−1,3-cyclohexanedione (NTBC) delays and in some cases fails to prevent disease progression to liver fibrosis, liver failure, and activation of tumorigenic pathways. Here we demonstrate cure of HT1 by direct, in vivo administration of a therapeutic lentiviral vector targeting the expression of a human fumarylacetoacetate hydrolase (FAH) transgene in the porcine model of HT1. This therapy is well tolerated and provides stable long-term expression of FAH in pigs with HT1. Genomic integration displays a benign profile, with subsequent fibrosis and tumorigenicity gene expression patterns similar to wild-type animals as compared to NTBC-treated or diseased untreated animals. Indeed, the phenotypic and genomic data following in vivo lentiviral vector administration demonstrate comparative superiority over other therapies including ex vivo cell therapy and therefore support clinical application of this approach. Hereditary tyrosinemia type 1 (HT1) is an inborn error of metabolism caused by a deficiency in fumarylacetoacetate hydrolase (FAH). Here, the authors show in an animal model that HT1 can be treated via in vivo portal vein administration of a lentiviral vector carrying the human FAH transgene.
Collapse
Affiliation(s)
- Clara T Nicolas
- Department of Surgery, Mayo Clinic, Rochester, MN, USA.,Faculty of Medicine, University of Barcelona, Barcelona, Spain.,Department of Surgery, University of Alabama Birmingham, Birmingham, AL, USA
| | | | - Raymond D Hickey
- Department of Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Zeji Du
- Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Lori G Hillin
- Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Rebekah M Guthman
- Department of Surgery, Mayo Clinic, Rochester, MN, USA.,Medical College of Wisconsin, Wausau, WI, USA
| | - William J Cao
- Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | | | | | - Diya Roy
- Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Aditya Bhagwate
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Daniel O'Brien
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Jean-Pierre Kocher
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Robert A Kaiser
- Department of Surgery, Mayo Clinic, Rochester, MN, USA.,Midwest Fetal Care Center, Children's Hospitals and Clinics of Minnesota, Minneapolis, MN, USA
| | | | - Joseph B Lillegard
- Department of Surgery, Mayo Clinic, Rochester, MN, USA. .,Midwest Fetal Care Center, Children's Hospitals and Clinics of Minnesota, Minneapolis, MN, USA. .,Pediatric Surgical Associates, Minneapolis, MN, USA.
| |
Collapse
|
11
|
Abbas K, Basit J, Rehman MEU. Adequacy of nitisinone for the management of alkaptonuria. Ann Med Surg (Lond) 2022; 80:104340. [PMID: 36045846 PMCID: PMC9422360 DOI: 10.1016/j.amsu.2022.104340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 07/28/2022] [Accepted: 07/31/2022] [Indexed: 11/24/2022] Open
Abstract
Alkaptonuria is a rare hereditary disease with a defective enzyme that results in increased homogentisic acid levels in the body. Homogentisic acid accumulates in multiple body parts and initializes tissue damage. Clinical manifestations such as pigmentation of the skin areas and joint destruction result in ochronosis. Nitisinone decreases serum and urinary homogentisic acid levels, improving morbidity by preventing and slowing the progression of alkaptonuria. Nitisinone-induced hypertyrosinemia causes keratopathy and mental ill effects, which can be managed by diet restriction and regular check-ups. A personalized approach is required for treatment by nitisinone. Low-dose oral nitisinone is associated with overall good results and a better safety profile.
Collapse
|
12
|
Menon J, Shanmugam N, Valamparampil JJ, Hakeem A, Vij M, Jalan A, Reddy MS, Rela M. Liver Transplantation: A Safe and Definitive Alternative to Lifelong Nitisinone for Tyrosinemia Type 1. Indian J Pediatr 2022; 89:438-444. [PMID: 34398413 DOI: 10.1007/s12098-021-03826-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 05/25/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVES To report the experience of liver transplantation (LT) for tyrosinemia type 1 (TT-1). METHODS Clinical data of children with TT-1 who underwent living donor LT between July 2009 and May 2020 were retrospectively analyzed. Data included pre-LT nitisinone therapy, graft type, post-LT complications, HCC incidence, and graft/patient survival. RESULTS Nine children were diagnosed with TT-1 at a median age of 12 mo (6-54 mo). Nitisinone was started in 6 patients at a median age of 15 mo (6-42 mo), but all had frequent interruption of therapy due to logistics with drug procurement including its cost. Median age at transplantation was 5 y (2-11 y). Explant liver showed HCC in 5 patients (55% of total cohort). The graft and patient survival are 100% with median follow-up of 58 mo (24-84 mo). CONCLUSION LT is curative for TT-1 and excellent results can be obtained in experienced centers. This is especially favorable in countries with limited resources where the cost of medical therapy is highly prohibitive, with lifelong diet restrictions and unclear long-term risk of HCC.
Collapse
Affiliation(s)
- Jagadeesh Menon
- Department of Pediatric Gastroenterology & Hepatology, Dr Rela Institute & Medical Centre, Bharat Institute of Higher Education and Research, Chennai, Tamil Nadu, 600044, India
| | - Naresh Shanmugam
- Department of Pediatric Gastroenterology & Hepatology, Dr Rela Institute & Medical Centre, Bharat Institute of Higher Education and Research, Chennai, Tamil Nadu, 600044, India.
| | - Joseph J Valamparampil
- Department of Pediatric Gastroenterology & Hepatology, Dr Rela Institute & Medical Centre, Bharat Institute of Higher Education and Research, Chennai, Tamil Nadu, 600044, India
| | - Abdul Hakeem
- Department of Hepatobiliary surgery & liver transplantation, Dr Rela Institute & Medical Centre, Bharat Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Mukul Vij
- Department of Histopathology, Dr Rela Institute & Medical Centre, Bharat Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Anil Jalan
- Department of Pediatric Genetics, NIRMAN, Mumbai, Maharashtra, India
| | - Mettu Srinivas Reddy
- Department of Hepatobiliary surgery & liver transplantation, Dr Rela Institute & Medical Centre, Bharat Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Mohamed Rela
- Department of Hepatobiliary surgery & liver transplantation, Dr Rela Institute & Medical Centre, Bharat Institute of Higher Education and Research, Chennai, Tamil Nadu, India.,Liver Transplant Unit, Kings College Hospital, London, UK
| |
Collapse
|
13
|
Davison AS, Hughes G, Harrold JA, Clarke P, Griffin R, Ranganath LR. Long-term low dose nitisinone therapy in adults with alkaptonuria shows no cognitive decline or increased severity of depression. JIMD Rep 2022; 63:221-230. [PMID: 35433173 PMCID: PMC8995840 DOI: 10.1002/jmd2.12272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/10/2022] [Accepted: 01/13/2022] [Indexed: 12/13/2022] Open
Abstract
Little is documented on whether nitisinone-induced hypertyrosinaemia alters cognitive functioning or leads to worsening depression in alkaptonuria (AKU). Wechsler Adult Intelligence Scale-IV (WAIS-IV) and Beck Depression Inventory-II (BDI-II) assessments were performed before and annually following treatment with nitisinone 2 mg daily to assess the impact on cognitive functioning and severity of depression. Serum tyrosine concentrations were also measured annually. WAIS-IV: 63 patients (27 females/36 males: mean age[years] [±standard deviation, range] 55.7[13.7, 26-79]; 60.3[9.6, 19-75]) were included at baseline for assessment of: verbal comprehension (VC), perceptual reasoning (PR), working memory (WM), and processing speed (PS) using separate indices. Over the 6-year period studied 43, 39, 36, 29, 26 and 15 patients had annual assessments. Using a longitudinal model (age and sex adjusted) no significant differences were observed in any of the indices over this period, apart from VC which showed a significant increase after adjustment for sex (p < 0.05). BDI-II: 74 patients (32 females/42 males: mean age[years] [±standard deviation, range] 56.1[13.2, 26-79]; 42 males, 51.5[16.3, 19-70]) were included at baseline. Over the 7-year period studied 48, 47, 38, 34, 32, 24 and 12 patients had annual assessments. No significant differences in BDI-II scores were observed when compared to baseline. Hypertyrosinaemia was observed in all patients following treatment with nitisinone (p < 0.001, at all annual visits). Serum tyrosine was not correlated with WAIS-IV sub-test indices or BDI-II scores pre- or post-nitisinone therapy. These findings suggest that treatment with nitisinone does not affect cognitive functioning and or lead to increased severity of depression.
Collapse
Affiliation(s)
- Andrew S. Davison
- Department of Clinical Biochemistry and Metabolic Medicine, Liverpool Clinical LaboratoriesRoyal Liverpool University HospitalLiverpoolUK
| | - Gin Hughes
- Department of PsychologyUniversity of LiverpoolLiverpoolUK
| | | | - Pam Clarke
- Department of PsychologyUniversity of LiverpoolLiverpoolUK
| | - Rebecca Griffin
- Liverpool Cancer Trials UnitUniversity of LiverpoolLiverpoolUK
| | - Lakshminarayan R. Ranganath
- Department of Clinical Biochemistry and Metabolic Medicine, Liverpool Clinical LaboratoriesRoyal Liverpool University HospitalLiverpoolUK
| |
Collapse
|
14
|
Kakos CD, Ziogas IA, Demiri CD, Esagian SM, Economopoulos KP, Moris D, Tsoulfas G, Alexopoulos SP. Liver Transplantation for Pediatric Hepatocellular Carcinoma: A Systematic Review. Cancers (Basel) 2022; 14:1294. [PMID: 35267604 PMCID: PMC8908995 DOI: 10.3390/cancers14051294] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 02/23/2022] [Accepted: 02/28/2022] [Indexed: 02/05/2023] Open
Abstract
Liver transplantation (LT) is the only potentially curative option for children with unresectable hepatocellular carcinoma (HCC). We performed a systematic review of the MEDLINE, Scopus, Cochrane Library, and Web of Science databases (end-of-search date: 31 July 2020). Our outcomes were overall survival (OS) and disease-free survival (DFS). We evaluated the effect of clinically relevant variables on outcomes using the Kaplan-Meier method and log-rank test. Sixty-seven studies reporting on 245 children undergoing LT for HCC were included. DFS data were available for 150 patients and the 1-, 3-, and 5-year DFS rates were 92.3%, 89.1%, and 84.5%, respectively. Sixty of the two hundred and thirty-eight patients (25.2%) died over a mean follow up of 46.8 ± 47.4 months. OS data were available for 222 patients and the 1-, 3-, and 5-year OS rates were 87.9%, 78.8%, and 74.3%, respectively. Although no difference was observed between children transplanted within vs. beyond Milan criteria (p = 0.15), superior OS was observed in children transplanted within vs. beyond UCSF criteria (p = 0.02). LT can yield favorable outcomes for pediatric HCC beyond Milan but not beyond UCSF criteria. Further research is required to determine appropriate LT selection criteria for pediatric HCC.
Collapse
Affiliation(s)
- Christos D. Kakos
- Surgery Working Group, Society of Junior Doctors, 15123 Athens, Greece; (C.D.K.); (I.A.Z.); (C.D.D.); (S.M.E.); (K.P.E.)
| | - Ioannis A. Ziogas
- Surgery Working Group, Society of Junior Doctors, 15123 Athens, Greece; (C.D.K.); (I.A.Z.); (C.D.D.); (S.M.E.); (K.P.E.)
- Department of Surgery, Division of Hepatobiliary Surgery and Liver Transplantation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Charikleia D. Demiri
- Surgery Working Group, Society of Junior Doctors, 15123 Athens, Greece; (C.D.K.); (I.A.Z.); (C.D.D.); (S.M.E.); (K.P.E.)
- 2nd Department of Pediatric Surgery, “Papageorgiou” General Hospital, Aristotle University School of Medicine, 54124 Thessaloniki, Greece
| | - Stepan M. Esagian
- Surgery Working Group, Society of Junior Doctors, 15123 Athens, Greece; (C.D.K.); (I.A.Z.); (C.D.D.); (S.M.E.); (K.P.E.)
| | - Konstantinos P. Economopoulos
- Surgery Working Group, Society of Junior Doctors, 15123 Athens, Greece; (C.D.K.); (I.A.Z.); (C.D.D.); (S.M.E.); (K.P.E.)
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA;
| | - Dimitrios Moris
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA;
| | - Georgios Tsoulfas
- Department of Surgery, Aristotle University School of Medicine, 54124 Thessaloniki, Greece;
| | - Sophoclis P. Alexopoulos
- Department of Surgery, Division of Hepatobiliary Surgery and Liver Transplantation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
15
|
Sikonja J, Brecelj J, Zerjav Tansek M, Repic Lampret B, Drole Torkar A, Klemencic S, Lipovec N, Stefanova Kralj V, Bertok S, Kovac J, Faganel Kotnik B, Tesarova M, Remec ZI, Debeljak M, Battelino T, Groselj U. Clinical and genetic characteristics of two patients with tyrosinemia type 1 in Slovenia – A novel fumarylacetoacetate hydrolase (FAH) intronic disease-causing variant. Mol Genet Metab Rep 2022; 30:100836. [PMID: 35242570 PMCID: PMC8856938 DOI: 10.1016/j.ymgmr.2021.100836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 11/02/2022] Open
Abstract
Tyrosinemia type 1 (HT1) is an inborn error of tyrosine catabolism that leads to severe liver, kidney, and neurological dysfunction. Newborn screening (NBS) can enable a timely diagnosis and early initiation of treatment. We presented the follow up of the only two Slovenian patients diagnosed with HT1. Metabolic control was monitored by measuring tyrosine, phenylalanine and succinylacetone from dried blood spots (DBSs). Retrograde screening of HT1 was performed from DBSs taken at birth using tandem mass spectrometry. First patient was diagnosed at the age of 6 months in the asymptomatic phase due to an abnormal liver echogenicity, the other presented at 2.5 months with an acute liver failure and needed a liver transplantation. The first was a compound heterozygote for a novel FAH intronic variant c.607-21A>G and c.192G>T whereas the second was homozygous for c.192G>T. At the non-transplanted patient, 66% of tyrosine and 79% of phenylalanine measurements were in strict reference ranges of 200–400 μmol/L and >30 μmol/L, respectively, which resulted in a favorable cognitive outcome at 3.6 years. On retrograde screening, both patients had elevated SA levels; on the other hand, tyrosine was elevated only at one. We showed that non-coding regions should be analyzed when clinical and biochemical markers are characteristic of HT1. DBSs represent a convenient sample type for frequent amino acid monitoring. Retrograde diagnosis of HT1 was possible after more than three years of birth with SA as a primary marker, complemented by tyrosine. Non-coding region variants of FAH gene can result in a symptomatic HT1. Retrograde screening for HT1 is technically possible even three years after birth. DBS are convenient for monitoring HT1 patients and are family-friendly. Regular monitoring in HT1 patients can result in a favorable cognitive outcome.
Collapse
|
16
|
Cannon Homaei S, Barone H, Kleppe R, Betari N, Reif A, Haavik J. ADHD symptoms in neurometabolic diseases: Underlying mechanisms and clinical implications. Neurosci Biobehav Rev 2021; 132:838-856. [PMID: 34774900 DOI: 10.1016/j.neubiorev.2021.11.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/09/2021] [Accepted: 11/09/2021] [Indexed: 12/16/2022]
Abstract
Neurometabolic diseases (NMDs) are typically caused by genetic abnormalities affecting enzyme functions, which in turn interfere with normal development and activity of the nervous system. Although the individual disorders are rare, NMDs are collectively relatively common and often lead to lifelong difficulties and high societal costs. Neuropsychiatric manifestations, including ADHD symptoms, are prominent in many NMDs, also when the primary biochemical defect originates in cells and tissues outside the nervous system. ADHD symptoms have been described in phenylketonuria, tyrosinemias, alkaptonuria, succinic semialdehyde dehydrogenase deficiency, X-linked ichthyosis, maple syrup urine disease, and several mitochondrial disorders, but are probably present in many other NMDs and may pose diagnostic and therapeutic challenges. Here we review current literature linking NMDs with ADHD symptoms. We cite emerging evidence that many NMDs converge on common neurochemical mechanisms that interfere with monoamine neurotransmitter synthesis, transport, metabolism, or receptor functions, mechanisms that are also considered central in ADHD pathophysiology and treatment. Finally, we discuss the therapeutic implications of these findings and propose a path forward to increase our understanding of these relationships.
Collapse
Affiliation(s)
- Selina Cannon Homaei
- Division of Psychiatry, Haukeland University Hospital, Norway; Department of Biomedicine, University of Bergen, Norway.
| | - Helene Barone
- Regional Resource Center for Autism, ADHD, Tourette Syndrome and Narcolepsy, Western Norway, Division of Psychiatry, Haukeland University Hospital, Norway.
| | - Rune Kleppe
- Division of Psychiatry, Haukeland University Hospital, Norway; Norwegian Centre for Maritime and Diving Medicine, Department of Occupational Medicine, Haukeland University Hospital, Norway.
| | - Nibal Betari
- Department of Biomedicine, University of Bergen, Norway.
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany.
| | - Jan Haavik
- Division of Psychiatry, Haukeland University Hospital, Norway; Department of Biomedicine, University of Bergen, Norway.
| |
Collapse
|
17
|
Casein Glycomacropeptide: An Alternative Protein Substitute in Tyrosinemia Type I. Nutrients 2021; 13:nu13093224. [PMID: 34579102 PMCID: PMC8467066 DOI: 10.3390/nu13093224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/08/2021] [Accepted: 09/14/2021] [Indexed: 12/25/2022] Open
Abstract
Tyrosinemia type I (HTI) is treated with nitisinone, a tyrosine (Tyr) and phenylalanine (Phe)-restricted diet, and supplemented with a Tyr/Phe-free protein substitute (PS). Casein glycomacropeptide (CGMP), a bioactive peptide, is an alternative protein source to traditional amino acids (L-AA). CGMP contains residual Tyr and Phe and requires supplementation with tryptophan, histidine, methionine, leucine, cysteine and arginine. Aims: a 2-part study assessed: (1) the tolerance and acceptability of a low Tyr/Phe CGMP-based PS over 28 days, and (2) its long-term impact on metabolic control and growth over 12 months. Methods: 11 children with HTI were recruited and given a low Tyr/Phe CGMP to supply all or part of their PS intake. At enrolment, weeks 1 and 4, caregivers completed a questionnaire on gastrointestinal symptoms, acceptability and ease of PS use. In study part 1, blood Tyr and Phe were assessed weekly; in part 2, weekly to fortnightly. In parts 1 and 2, weight and height were assessed at the study start and end. Results: Nine of eleven children (82%), median age 15 years (range 8.6–17.7), took low Tyr/Phe CGMP PS over 28 days; it was continued for 12 months in n = 5 children. It was well accepted by 67% (n = 6/9), tolerated by 100% (n = 9/9) and improved gastrointestinal symptoms in 2 children. The median daily dose of protein equivalent from protein substitute was 60 g/day (range 45–60 g) with a median of 20 g/day (range 15 to 30 g) from natural protein. In part 2 (n = 5), a trend for improved blood Tyr was observed: 12 months pre-study, median Tyr was 490 μmol/L (range 200–600) and Phe 50 μmol/L (range 30–100); in the 12 months taking low Tyr/Phe CGMP PS, median Tyr was 430 μmol/L (range 270–940) and Phe 40 μmol/L (range 20–70). Normal height, weight and BMI z scores were maintained over 12 months. Conclusions: In HTI children, CGMP was well tolerated, with no deterioration in metabolic control or growth when studied over 12 months. The efficacy of CGMP in HTI needs further investigation to evaluate the longer-term impact on blood Phe concentrations and its potential influence on gut microflora
Collapse
|
18
|
Yilmaz O, Daly A, Pinto A, Ashmore C, Evans S, Gupte G, Jackson R, Yabanci Ayhan N, MacDonald A. Physical Growth of Patients with Hereditary Tyrosinaemia Type I: A Single-Centre Retrospective Study. Nutrients 2021; 13:3070. [PMID: 34578949 PMCID: PMC8472760 DOI: 10.3390/nu13093070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/26/2021] [Accepted: 08/30/2021] [Indexed: 12/23/2022] Open
Abstract
In a retrospective review, we aimed to assess long-term growth in 17 patients (n = 11 males) with hereditary tyrosinaemia type I (HTI). Median age at assessment was 15.6 years (5.7-26.6 years) and median age at diagnosis was 1 month (range: 0-16 months), with 35% (n = 6/17) symptomatic on presentation. From the age of 8 years, there was a noticeable change in median height, weight, and body-mass-index [BMI]-z-scores. Median height-for-age z-scores were consistently ≤ -1 (IQR -1.6, -0.5) during the first 8 years of life but increased with age. Weight-for-age z-scores ranged between -1 to 0 (IQR -1.2, 0.1) in the first 8 years; then increased to > 0.5 (IQR -0.3, 1.3) by age 16 years, and BMI-for-age z-scores ranged from 0 to 1 (IQR -0.7, 1.3) up to 8 years, and >1 (IQR -0.2, 1.9) until 16 years. The percentage of overweight and obesity was lowest in children aged < 5 years, and consistently > 40% in patients aged between 7 to 16 years. The prescribed total protein intake was associated with improved height growth (p < 0.01). Impaired growth in early life improved with age achieving normal population standards. Further studies are needed to investigate factors that influence growth outcome in HTI patients.
Collapse
Affiliation(s)
- Ozlem Yilmaz
- Birmingham Women’s and Children’s Hospital, Birmingham B4 6NH, UK; (O.Y.); (A.D.); (A.P.); (C.A.); (S.E.); (G.G.)
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Ankara Yildirim Beyazit University, Ankara 06760, Turkey
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Ankara University, Ankara 06290, Turkey;
| | - Anne Daly
- Birmingham Women’s and Children’s Hospital, Birmingham B4 6NH, UK; (O.Y.); (A.D.); (A.P.); (C.A.); (S.E.); (G.G.)
| | - Alex Pinto
- Birmingham Women’s and Children’s Hospital, Birmingham B4 6NH, UK; (O.Y.); (A.D.); (A.P.); (C.A.); (S.E.); (G.G.)
| | - Catherine Ashmore
- Birmingham Women’s and Children’s Hospital, Birmingham B4 6NH, UK; (O.Y.); (A.D.); (A.P.); (C.A.); (S.E.); (G.G.)
| | - Sharon Evans
- Birmingham Women’s and Children’s Hospital, Birmingham B4 6NH, UK; (O.Y.); (A.D.); (A.P.); (C.A.); (S.E.); (G.G.)
| | - Girish Gupte
- Birmingham Women’s and Children’s Hospital, Birmingham B4 6NH, UK; (O.Y.); (A.D.); (A.P.); (C.A.); (S.E.); (G.G.)
| | - Richard Jackson
- Cancer Research UK Liverpool Cancer Trials Unit, University of Liverpool, Liverpool L69 3GL, UK;
| | - Nurcan Yabanci Ayhan
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Ankara University, Ankara 06290, Turkey;
| | - Anita MacDonald
- Birmingham Women’s and Children’s Hospital, Birmingham B4 6NH, UK; (O.Y.); (A.D.); (A.P.); (C.A.); (S.E.); (G.G.)
| |
Collapse
|
19
|
Daou KN, Barhoumi A, Bassyouni A, Karam PE. Diagnostic and Therapeutic Challenges of Hereditary Tyrosinemia Type 1 in Lebanon: A 12-Year Retrospective Review. Front Pediatr 2021; 9:698577. [PMID: 34422723 PMCID: PMC8377248 DOI: 10.3389/fped.2021.698577] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/06/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Hereditary tyrosinemia type 1 is a rare genetic disorder leading to liver cirrhosis and hepatocellular carcinoma. Few decades ago, dietary measures and ultimately liver transplant constituted the only treatment modalities. Nowadays, early diagnosis and therapy with nitisinone can reverse the clinical picture. In developing countries, diagnostic and therapeutic challenges may affect the outcome of this disease. The choice of the treatment modality may depend on the economic status of each country. Few reports on the long-term outcome of hereditary tyrosinemia type 1 are available from developing and Arab countries. Methods: A retrospective study of charts of Lebanese patients diagnosed with tyrosinemia type 1 and followed, at the American University of Beirut, during a 12-year period was performed. Clinical presentation and liver biochemical profile at diagnosis were analyzed, along with therapeutic modalities and long-term outcome. Results: Twenty-two children were diagnosed and followed during the study period. Median age at diagnosis was 7 months (range: one day to 35 months). Most of the patients presented with hepatomegaly and jaundice. Four patients were referred for atypical presentations with developmental delay and seizures, secondary to undiagnosed hypoglycemia episodes. Around half of the patients presented with failure to thrive. Transaminitis, cholestasis and increased α-fetoprotein level were variably present at diagnosis (36% to 50%). All patients had elevated plasma tyrosine and urinary succinylacetone levels. Genetic testing was performed in 9%. Only one third could be treated with nitisinone. Liver transplant was electively performed in 9% of cases, to overcome the long-term cost of nitisinone. One third of the patients died between the age of 1 month and 11 years. Surviving patients are still candidates for liver transplant. Conclusion: Our experience reflects the challenges of diagnosis and treatment of hereditary tyrosinemia type 1 in a developing country. In the absence of specific neonatal screening, early diagnosis relies mostly on the clinical awareness of the physician. Long-term nitisinone use may be deterred by its high cost and liver transplantation carries risks of surgical complications. New, effective, and less expensive treatments are needed, especially for developing countries.
Collapse
Affiliation(s)
- Karim N. Daou
- Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Abir Barhoumi
- Department of Nutrition, American University of Beirut Medical Center, Beirut, Lebanon
- Inherited Metabolic Diseases Program, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Amina Bassyouni
- Inherited Metabolic Diseases Program, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Pascale E. Karam
- Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Inherited Metabolic Diseases Program, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
20
|
Spiekerkoetter U, Couce ML, Das AM, de Laet C, Dionisi-Vici C, Lund AM, Schiff M, Spada M, Sparve E, Szamosi J, Vara R, Rudebeck M. Long-term safety and outcomes in hereditary tyrosinaemia type 1 with nitisinone treatment: a 15-year non-interventional, multicentre study. Lancet Diabetes Endocrinol 2021; 9:427-435. [PMID: 34023005 DOI: 10.1016/s2213-8587(21)00092-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/24/2021] [Accepted: 03/30/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Since the EU approval of nitisinone in 2005, prognosis for patients with hereditary tyrosinaemia type 1 has changed dramatically, with patients living with the disease now reaching adulthood for the first time in history. This study aimed to assess the long-term safety and outcomes of nitisinone treatment in patients with hereditary tyrosinaemia type 1. METHODS We did a non-interventional, non-comparative, multicentre study in 77 sites across 17 countries in Europe and collected retrospective and prospective longitudinal data in patients with hereditary tyrosinaemia type 1 who were treated with oral nitisinone during the study period (Feb 21, 2005, to Sept 30, 2019). There were no specific exclusion criteria. Patients were followed-up with an investigator at least annually for as long as they were treated, or until the end of the study. The primary endpoints, occurrence of adverse events related to hepatic, renal, ophthalmic, haematological, or cognitive or developmental function, were assessed in the complete set (all patients already receiving treatment at the index date [Feb 21, 2005] or starting treatment thereafter) and the index set (the subset of patients who had their first dose on the index date or later only). FINDINGS 315 patients were enrolled during the study period (complete set). Additionally, data from 24 patients who had liver transplantation or died during the post-marketing surveillance programme were retrieved (extended analysis set; 339 patients). Median treatment duration was 11·2 years (range 0·7-28·4); cumulative nitisinone exposure was 3172·7 patient-years. Patients who were diagnosed by neonatal screening started nitisinone treatment at median age 0·8 months versus 8·5 months in those who presented clinically. Incidences of hepatic, renal, ophthalmic, haematological, or cognitive or developmental adverse events were low. Occurrence of liver transplantation or death was more frequent the later that treatment was initiated (none of 70 patients who started treatment at age <28 days vs 35 [13%] of 268 patients who started treatment at age ≥28 days). 279 (89%) of 315 patients were assessed as having either very good or good nitisinone treatment compliance. Treatment and diet compliance declined as patients aged. Suboptimal plasma phenylalanine and tyrosine levels were observed. The majority of patients were reported to have good overall clinical condition throughout treatment; 176 (87%) of 203 during the entire study, 98% following 1 year of treatment. INTERPRETATION Long-term nitisinone treatment was well tolerated and no new safety signals were revealed. Life-limiting hepatic disease appears to have been prevented by early treatment start. Neonatal screening was the most effective way of ensuring early treatment. Standardised monitoring of blood tyrosine, phenylalanine, and nitisinone levels has potential to guide individualised therapy. FUNDING Swedish Orphan Biovitrum (Sobi).
Collapse
Affiliation(s)
- Ute Spiekerkoetter
- Department of Paediatrics and Adolescent Medicine, University Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Maria L Couce
- Hospital Clínico Universitario de Santiago de Compostela, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), MetabERN, Santiago de Compostela, Spain
| | - Anibh M Das
- Department of Paediatrics, Hannover Medical School, Hannover, Germany
| | - Corinne de Laet
- Nutrition and Metabolism Unit, Department of Paediatrics, Hôpital Universitaire des Enfants Reine Fabiola, Brussels, Belgium
| | - Carlo Dionisi-Vici
- Division of Metabolism, Bambino Gesù Children's Hospital Instituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Allan M Lund
- Centre for Inherited Metabolic Diseases, Departments of Paediatrics and Clinical Genetics, Copenhagen University Hospital, Copenhagen, Denmark
| | - Manuel Schiff
- Necker Hospital, AP-HP, Reference Centre for Inborn Error of Metabolism (Filière G2M), Paediatrics Department, University of Paris, Paris, France; Inserm UMR_S1163, Institut Imagine, Paris, France
| | - Marco Spada
- Department of Paediatrics, Regina Margherita Children Hospital, University of Torino, Torino, Italy
| | - Erik Sparve
- Swedish Orphan Biovitrum (Sobi), Stockholm, Sweden
| | | | - Roshni Vara
- Department of Paediatric Inherited Metabolic Disease, Evelina London Children's Hospital, London, UK
| | | |
Collapse
|
21
|
Treatment adherence in tyrosinemia type 1 patients. Orphanet J Rare Dis 2021; 16:256. [PMID: 34082789 PMCID: PMC8173906 DOI: 10.1186/s13023-021-01879-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 05/21/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND While therapeutic advances have significantly improved the prognosis of patients with hereditary tyrosinemia type 1 (HT1), adherence to dietary and pharmacological treatments is essential for an optimal clinical outcome. Poor treatment adherence is well documented among patients with chronic diseases, but data from HT1 patients are scarce. This study evaluated pharmacological and dietary adherence in HT1 patients both directly, by quantifying blood levels nitisinone (NTBC) levels and metabolic biomarkers of HT1 [tyrosine (Tyr), phenylalanine (Phe), and succinylacetone]; and indirectly, by analyzing NTBC prescriptions from hospital pharmacies and via clinical interviews including the Haynes-Sackett (or self-compliance) test and the adapted Battle test of patient knowledge of the disease. RESULTS This observational study analyzed data collected over 4 years from 69 HT1 patients (7 adults and 62 children; age range, 7 months-35 years) who were treated with NTBC and a low-Tyr, low-Phe diet. Adherence to both pharmacological and, in particular, dietary treatment was poor. Annual data showed that NTBC levels were lower than recommended in more than one third of patients, and that initial Tyr levels were high (> 400 µM) in 54.2-64.4% of patients and exceeded 750 µM in 25.8% of them. Remarkably, annual normalization of NTBC levels was observed in 29.4-57.9% of patients for whom serial NTBC determinations were performed. Poor adherence to dietary treatment was more refractory to positive reinforcement: 36.2% of patients in the group who underwent multiple analyses per year maintained high Tyr levels during the entire study period, and, when considering each of the years individually this percentage ranged from 75 to 100% of them. Indirect methods revealed percentages of non-adherent patients of 7.3 and 15.9% (adapted Battle and Haynes tests, respectively). CONCLUSIONS Despite initially poor adherence to pharmacological and especially dietary treatment among HT1 patients, positive reinforcement at medical consultations resulted in a marked improvement in NTBC levels, indicating the importance of systematic positive reinforcement at medical visits.
Collapse
|
22
|
Dweikat I, Qawasmi N, Najeeb A, Radwan M. Phenotype, genotype, and outcome of 25 Palestinian patients with hereditary tyrosinemia type 1. Metabol Open 2021; 9:100083. [PMID: 33598652 PMCID: PMC7868710 DOI: 10.1016/j.metop.2021.100083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/22/2021] [Accepted: 01/23/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Tyrosinemia type 1 (hepatorenal tyrosinemia, HT1) is a rare autosomal recessive inborn error of tyrosine metabolism caused by deficiency of the last enzyme in the tyrosine catabolic pathway, fumarylacetoacetate hydrolase (FAH) leading to severe hepatic, renal and peripheral nerve damage if left untreated. Early treatment may prevent acute liver failure, renal dysfunction, liver cirrhosis, hepatocellular carcinoma (HCC) and improves survival. MATERIAL AND METHODS A retrospective single center study was carried out based on the clinical and biochemical presentation, therapy and outcome of 25 Palestinian patients with HT1 diagnosed during the last 25 years. RESULTS HT1 is not included in newborn screening program in Palestine. The mean age at diagnosis was 8 months and the main clinical manifestations were coagulopathy, hepatomegaly, splenomegaly and renal tubular dysfunction. The main biochemical abnormalities were elevated plasma tyrosine, serum transaminases and prothrombin time, and low serum phosphorous with elevated alkaline phosphatase compatible with hypophosphatemic rickets secondary to renal tubular dysfunction. All patients were treated with nitisinone. The mean duration of nitisinone treatment was 74 months and the mean dosage was 0.89 mg/kg/day. None developed HCC or neurological crisis. CONCLUSIONS Most patients present with liver failure and renal tubular dysfunction. Nitisinone treatment was effective therapy in all patients and improved both short- and long-term prognosis of HT1. Renal tubular dysfunction improved in all patients within the first week of starting nitisinone. Early diagnosis is necessary because delay in the treatment increases the risk of progressive liver failure HCC, progressive renal disease and neuropathy.
Collapse
Affiliation(s)
- Imad Dweikat
- Metabolic Department Arab American University, PO Box 240 Jenin, 13 Zababdeh, Palestine
| | - Nada Qawasmi
- Pediatric Department, Makassed Hospital, Palestine
| | - Aysha Najeeb
- Pediatric Department, Makassed Hospital, Palestine
| | | |
Collapse
|
23
|
Ray SK, Mukherjee S. Molecular and biochemical investigations of inborn errors of metabolism-altered redox homeostasis in branched-chain amino acid disorders, organic acidurias, and homocystinuria. Free Radic Res 2021; 55:627-640. [PMID: 33504220 DOI: 10.1080/10715762.2021.1877286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
India, resembling other developing nations, is confronting a hastening demographic switch to non-communicable diseases. Inborn errors of metabolism (IEM) constitute a varied heterogeneous group of disorders with variable clinical appearance, primarily in the pediatric populace. Congenital deformities and genetic disorders are significant for mortality throughout the world, and the Indian scenario is not very different. IEMs are a group of monogenic issues described by dysregulation of the metabolic networks that bring about development and homeostasis. Incipient evidence focuses on oxidative stress and mitochondrial dysfunction as significant contributors to the multiorgan modifications are detected in a few IEMs. The amassing of toxic metabolites in organic acidurias, respiratory chain, and fatty acid oxidation ailments inhibit mitochondrial enzymes and processes, bringing about elevated levels of reactive oxygen species (ROS). In different IEMs, as in homocystinuria, various sources of ROS have been suggested. In patients' samples along with cellular and experimental animal models, a few investigations have recognized substantial increments in ROS levels alongside diminishes in antioxidant defenses, relating with oxidative damage to proteins, lipids as well as DNA. Elevated ROS levels interrupt redox signaling pathways controlling biological processes such as cell development, differentiation, or apoptosis; however, few investigations explore these processes in IEMs. This review depicts the mitochondrial dysfunction, oxidative stress, redox signaling in branched-chain amino acid disorders, further organic acidurias, and homocystinuria, alongside the latest research investigating the proficiency of antioxidants in addition to mitochondria-targeted therapies as therapeutic components in these diseases.
Collapse
Affiliation(s)
- Suman Kumar Ray
- Department of Applied Sciences, Indira Gandhi Technological and Medical Sciences University, Ziro, Arunachal , Pradesh, India
| | - Sukhes Mukherjee
- Department of Biochemistry, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| |
Collapse
|
24
|
Mirani S, Poojari V, Shetty NS, Shah I. Outcome of Tyrosinemia Type 1 in Indian Children. J Clin Exp Hepatol 2021; 11:9-13. [PMID: 33679043 PMCID: PMC7897851 DOI: 10.1016/j.jceh.2020.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 07/05/2020] [Indexed: 12/12/2022] Open
Abstract
AIM The objective of this study was to determine the outcome of children with tyrosinemia type 1 from India. METHODS A retrospective observational study was conducted on 11 patients diagnosed with type I tyrosinemia under our care. Age at symptoms, age at diagnosis, age at starting 2-nitro-4-trifluoromethylbenzoyl-1,3-cyclohexanedione (NTBC), duration between diagnosis and initiation of NTBC, dose given, total duration of NTBC, and outcomes were noted. RESULTS Eleven children with a median age of 1.1 years (0.51-1.52) at onset of symptoms were included in the study. The median age at diagnosis was 1.76 years (0.95-2.43). Their current median age is 5.44 (2.36-8.80) years. Common clinical features at presentation were chronic liver disease in 8 (72.72%), rickets in 2 (18.18%), and fulminant liver disease in 1 (9.09%) patient. Hepatomegaly was observed in all children, growth retardation in 9 (81.81%), coagulopathy in 8 (72.72%), and abdominal distention in 6 (54.54%) patients. The median duration of NTBC therapy was 13.5 (7-21.25) months. The median dose of NTBC was 1 (0.77-1) mg/kg/day. One (9.09%) patient died due to liver cell failure. However, she had received NTBC only for a month. Another patient developed hepatocellular carcinoma (HCC) and underwent liver transplantation. He could receive NTBC only for 2 months, although he was diagnosed to have tyrosinemia for over a 1 year. Eight patients are on treatment with NTBC and are doing well, and 1 patient is not on NTBC and continues to have renal tubular acidosis. CONCLUSION NTBC therapy is effective and improves the prognosis of tyrosinemia. A long-term follow-up is required to determine progression to HCC and need for liver transplantation.
Collapse
Key Words
- AFP, Alpha-feto Protein
- ALP, Alkaline Phosphatase
- CLD, Chronic Liver Disease
- GGPT, γ-glutamine Transaminase
- HCC, Hepatocellular Carcinoma
- INR, International Normalized Ratio
- LFT, Liver Function Test
- NTBC
- NTBC, 2-nitro-4-trifluoromethylbenzoyl-1,3-cyclohexanedione
- RTA, Renal Tubular Acidosis
- SA, Succinylacetone
- SGOT, Aspartate Transaminase
- SGPT, Alanine Transaminase
- Tyrosinemia type 1
- hepatocellular carcinoma
- liver transplant
- succinylacetone
Collapse
Affiliation(s)
- Sonal Mirani
- Department of Pediatric Gastroenterology and Hepatology, B J Wadia Hospital for Children, Mumbai, India
| | - Vishrutha Poojari
- Department of Pediatric Gastroenterology and Hepatology, B J Wadia Hospital for Children, Mumbai, India
| | - Naman S. Shetty
- Department of Pediatric Gastroenterology and Hepatology, B J Wadia Hospital for Children, Mumbai, India
| | - Ira Shah
- Department of Pediatric Gastroenterology and Hepatology, B J Wadia Hospital for Children, Mumbai, India,Consultant in Pediatric Gastroenterology and Hepatology, Nanavati Hospital, Mumbai, India,Address for correspondence. Ira Shah, Head, Department of Pediatric Gastroenterology and Hepatology, B J Wadia Hospital for Children, Mumbai, India.
| |
Collapse
|
25
|
Bharadwaj A, Wahi N, Saxena A. Occurrence of Inborn Errors of Metabolism in Newborns, Diagnosis and Prophylaxis. Endocr Metab Immune Disord Drug Targets 2020; 21:592-616. [PMID: 33357204 DOI: 10.2174/1871530321666201223110918] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/09/2020] [Accepted: 11/09/2020] [Indexed: 11/22/2022]
Abstract
Inborn errors of metabolism (IEM) are a heterogeneous group of rare genetic disorders that are generally transmitted as autosomal or X-linked recessive disorders. These defects arise due to mutations associated with specific gene(s), especially the ones associated with key metabolic enzymes. These enzymes or their product(s) are involved in various metabolic pathways, leading to the accumulation of intermediary metabolite(s), reflecting their toxic effects upon mutations. The diagnosis of these metabolic disorders is based on the biochemical analysis of the clinical manifestations produced and their molecular mechanism. Therefore, it is imperative to devise diagnostic tests with high sensitivity and specificity for early detection of IEM. Recent advances in biochemical and polymerase chain reaction-based genetic analysis along with pedigree and prenatal diagnosis can be life-saving in nature. The latest development in exome sequencing for rapid diagnosis and enzyme replacement therapy would facilitate the successful treatment of these metabolic disorders in the future. However, the longterm clinical implications of these genetic manipulations is still a matter of debate among intellectuals and requires further research.
Collapse
Affiliation(s)
- Alok Bharadwaj
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Nitin Wahi
- Department of Bioinformatics, Pathfinder Research and Training Foundation, Greater Noida - 201308, Uttar Pradesh, India
| | - Aditya Saxena
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| |
Collapse
|
26
|
Colemonts-Vroninks H, Neuckermans J, Marcelis L, Claes P, Branson S, Casimir G, Goyens P, Martens GA, Vanhaecke T, De Kock J. Oxidative Stress, Glutathione Metabolism, and Liver Regeneration Pathways Are Activated in Hereditary Tyrosinemia Type 1 Mice upon Short-Term Nitisinone Discontinuation. Genes (Basel) 2020; 12:E3. [PMID: 33375092 PMCID: PMC7822164 DOI: 10.3390/genes12010003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/14/2022] Open
Abstract
Hereditary tyrosinemia type 1 (HT1) is an inherited condition in which the body is unable to break down the amino acid tyrosine due to mutations in the fumarylacetoacetate hydrolase (FAH) gene, coding for the final enzyme of the tyrosine degradation pathway. As a consequence, HT1 patients accumulate toxic tyrosine derivatives causing severe liver damage. Since its introduction, the drug nitisinone (NTBC) has offered a life-saving treatment that inhibits the upstream enzyme 4-hydroxyphenylpyruvate dioxygenase (HPD), thereby preventing production of downstream toxic metabolites. However, HT1 patients under NTBC therapy remain unable to degrade tyrosine. To control the disease and side-effects of the drug, HT1 patients need to take NTBC as an adjunct to a lifelong tyrosine and phenylalanine restricted diet. As a consequence of this strict therapeutic regime, drug compliance issues can arise with significant influence on patient health. In this study, we investigated the molecular impact of short-term NTBC therapy discontinuation on liver tissue of Fah-deficient mice. We found that after seven days of NTBC withdrawal, molecular pathways related to oxidative stress, glutathione metabolism, and liver regeneration were mostly affected. More specifically, NRF2-mediated oxidative stress response and several toxicological gene classes related to reactive oxygen species metabolism were significantly modulated. We observed that the expression of several key glutathione metabolism related genes including Slc7a11 and Ggt1 was highly increased after short-term NTBC therapy deprivation. This stress response was associated with the transcriptional activation of several markers of liver progenitor cells including Atf3, Cyr61, Ddr1, Epcam, Elovl7, and Glis3, indicating a concreted activation of liver regeneration early after NTBC withdrawal.
Collapse
Affiliation(s)
- Haaike Colemonts-Vroninks
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium; (H.C.-V.); (J.N.); (P.C.); (S.B.); (T.V.)
| | - Jessie Neuckermans
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium; (H.C.-V.); (J.N.); (P.C.); (S.B.); (T.V.)
| | - Lionel Marcelis
- Laboratoire de Pédiatrie, Hôpital Universitaire des Enfants Reine Fabiola (HUDERF), Université Libre de Bruxelles (ULB), Avenue J.J. Crocq 1-3, 1020 Brussels, Belgium; (L.M.); (G.C.); (P.G.)
| | - Paul Claes
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium; (H.C.-V.); (J.N.); (P.C.); (S.B.); (T.V.)
| | - Steven Branson
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium; (H.C.-V.); (J.N.); (P.C.); (S.B.); (T.V.)
| | - Georges Casimir
- Laboratoire de Pédiatrie, Hôpital Universitaire des Enfants Reine Fabiola (HUDERF), Université Libre de Bruxelles (ULB), Avenue J.J. Crocq 1-3, 1020 Brussels, Belgium; (L.M.); (G.C.); (P.G.)
| | - Philippe Goyens
- Laboratoire de Pédiatrie, Hôpital Universitaire des Enfants Reine Fabiola (HUDERF), Université Libre de Bruxelles (ULB), Avenue J.J. Crocq 1-3, 1020 Brussels, Belgium; (L.M.); (G.C.); (P.G.)
| | - Geert A. Martens
- Department of Laboratory Medicine, AZ Delta General Hospital, Deltalaan 1, 8800 Roeselare, Belgium;
- Center for Beta Cell Therapy in Diabetes, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Tamara Vanhaecke
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium; (H.C.-V.); (J.N.); (P.C.); (S.B.); (T.V.)
| | - Joery De Kock
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium; (H.C.-V.); (J.N.); (P.C.); (S.B.); (T.V.)
| |
Collapse
|
27
|
Abstract
Background Introduction of nitisinone and newborn screening (NBS) have transformed the treatment of type 1 tyrosinemia, but the effects of these changes on the long-term outcomes remain obscure. Also, the predictors for later complications, the significance of drug levels and the normalization of laboratory and imaging findings are poorly known. We investigated these issues in a nationwide study. Results Type 1 tyrosinemia was diagnosed in 22 children in 1978–2019 in Finland. Incidence was 1/90,102, with a significant enrichment in South Ostrobothnia (1/9990). Median age at diagnosis was 5 (range 0.5–36) months, 55% were girls and 13 had homozygotic Trp262X mutation. Four patients were detected through screening and 18 clinically, their main findings being liver failure (50% vs. 100%, respectively, p = 0.026), ascites (0% vs. 53%, p = 0.104), renal tubulopathy (0% vs. 65%, p = 0.035), rickets (25% vs. 65%, p = 0.272), growth failure (0% vs. 66%, p = 0.029), thrombocytopenia (25% vs. 88%, p = 0.028) and anaemia (0% vs. 47%, p = 0.131). One patient was treated with diet, seven with transplantation and 14 with nitisinone. Three late-diagnosed (6–33 months) nitisinone treated patients needed transplantation later. Kidney dysfunction (86% vs. 7%, p = 0.001), hypertension (57% vs. 7%, p = 0.025) and osteopenia/osteoporosis (71% vs. 14%, p = 0.017) were more frequent in transplanted than nitisinone-treated patients. Blood/serum alpha-fetoprotein decreased rapidly on nitisinone in all but one patient, who later developed intrahepatic hepatocellular carcinoma. Liver values normalized in 31 months and other laboratory values except thrombocytopenia within 18 months. Imaging findings normalized in 3–56 months excluding five patients with liver or splenic abnormalities. Low mean nitisinone concentration was associated with higher risk of severe complications (r = 0.758, p = 0.003) despite undetectable urine succinylacetone. Conclusions Prognosis of type 1 tyrosinemia has improved in the era of nitisinone, and NBS seems to provide further benefits. Nevertheless, the long-term risk for complications remains, particularly in the case of late diagnosis and/or insufficient nitisinone levels.
Collapse
|
28
|
Schultz MJ, Netzel BC, Singh RH, Pino GB, Gavrilov DK, Oglesbee D, Raymond KM, Rinaldo P, Tortorelli S, Smith WE, Matern D. Laboratory monitoring of patients with hereditary tyrosinemia type I. Mol Genet Metab 2020; 130:247-254. [PMID: 32546364 DOI: 10.1016/j.ymgme.2020.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 05/31/2020] [Accepted: 06/01/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND The prognosis of patients with Hereditary Tyrosinemia Type 1 (HT-1) has greatly improved with early detection through newborn screening and the introduction of nitisinone (NTBC) therapy. A recent guideline calls for periodic monitoring of biochemical markers and NTBC levels to tailor treatment; however, this is currently only achieved through a combination of clinical laboratory tests. We developed a multiplexed assay measuring relevant amino acids, succinylacetone (SUAC), and NTBC in dried blood spots (DBS) to facilitate treatment monitoring. METHODS Tyrosine, phenylalanine, methionine, NTBC and SUAC were eluted from DBS with methanol containing internal standards for each analyte and analyzed by liquid chromatography tandem mass spectrometry over 6.5 min in the multiple reaction monitoring positive mode. RESULTS Pre-analytical and analytical factors were studied and demonstrated a reliable assay. Chromatography resolved an unknown substance that falsely elevates SUAC concentrations and was present in all samples. To establish control and disease ranges, the method was applied to DBS collected from controls (n = 284) and affected patients before (n = 2) and after initiation of treatment (n = 29). In the treated patients SUAC concentrations were within the normal range over a wide range of NTBC levels. CONCLUSIONS This assay enables combined, accurate measurement of revelevant metabolites and NTBC in order to simplify treatment monitoring of patients with HT-1. In addition, the use of DBS allows for specimen collection at home to facilitate more standardization in relation to drug and dietary treatment.
Collapse
Affiliation(s)
- Matthew J Schultz
- Biochemical Genetics Laboratory, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Brian C Netzel
- Biochemical Genetics Laboratory, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Rani H Singh
- Department of Human Genetics and Pediatrics, Emory University, Atlanta, GA, USA
| | - Gisele B Pino
- Biochemical Genetics Laboratory, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Dimitar K Gavrilov
- Biochemical Genetics Laboratory, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Devin Oglesbee
- Biochemical Genetics Laboratory, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Kimiyo M Raymond
- Biochemical Genetics Laboratory, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Piero Rinaldo
- Biochemical Genetics Laboratory, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Silvia Tortorelli
- Biochemical Genetics Laboratory, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Wendy E Smith
- Maine Medical Partners Pediatrics Specialty Care, Portland, ME, USA
| | - Dietrich Matern
- Biochemical Genetics Laboratory, Mayo Clinic College of Medicine, Rochester, MN, USA.
| |
Collapse
|
29
|
Thompson WS, Mondal G, Vanlith CJ, Kaiser RA, Lillegard JB. The future of gene-targeted therapy for hereditary tyrosinemia type 1 as a lead indication among the inborn errors of metabolism. Expert Opin Orphan Drugs 2020; 8:245-256. [PMID: 33224636 PMCID: PMC7676758 DOI: 10.1080/21678707.2020.1791082] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction Inborn errors of metabolism (IEMs) often result from single-gene mutations and collectively cause liver dysfunction in neonates leading to chronic liver and systemic disease. Current treatments for many IEMs are limited to maintenance therapies that may still require orthotropic liver transplantation. Gene therapies offer a potentially superior approach by correcting or replacing defective genes with functional isoforms; however, they face unique challenges from complexities presented by individual diseases and their diverse etiology, presentation, and pathophysiology. Furthermore, immune responses, off-target gene disruption, and tumorigenesis are major concerns that need to be addressed before clinical application of gene therapy. Areas covered The current treatments for IEMs are reviewed as well as the advances in, and barriers to, gene therapy for IEMs. Attention is then given to ex vivo and in vivo gene therapy approaches for hereditary tyrosinemia type 1 (HT1). Of all IEMs, HT1 is particularly amenable to gene therapy because of a selective growth advantage conferred to corrected cells, thereby lowering the initial transduction threshold for phenotypic relevance. Expert opinion It is proposed that not only is HT1 a safe indication for gene therapy, its unique characteristics position it to be an ideal IEM to develop for clinical investigation.
Collapse
Affiliation(s)
| | - Gourish Mondal
- Department of Surgery, Research Scientist, Mayo Clinic, Rochester, MN, USA
| | | | - Robert A Kaiser
- Department of Surgery, Research Scientist, Mayo Clinic, Rochester, MN, USA.,Midwest Fetal Care Center, Childrens Hospital of Minnesota, MN, USA
| | - Joseph B Lillegard
- Midwest Fetal Care Center, Childrens Hospital of Minnesota, MN, USA.,Assistant Professor of Surgery, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
30
|
Priestley JRC, Alharbi H, Callahan KP, Guzman H, Payan-Walters I, Smith L, Ficicioglu C, Ganetzky RD, Ahrens-Nicklas RC. The Importance of Succinylacetone: Tyrosinemia Type I Presenting with Hyperinsulinism and Multiorgan Failure Following Normal Newborn Screening. Int J Neonatal Screen 2020; 6:39. [PMID: 32832707 PMCID: PMC7422996 DOI: 10.3390/ijns6020039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/14/2020] [Indexed: 11/16/2022] Open
Abstract
Tyrosinemia type 1 (TT1) is an inborn error of tyrosine metabolism with features including liver dysfunction, cirrhosis, and hepatocellular carcinoma; renal dysfunction that may lead to failure to thrive and bone disease; and porphyric crises. Once fatal in most infantile-onset cases, pre-symptomatic diagnosis through newborn screening (NBS) protocols, dietary management, and pharmacotherapy with nitisinone have improved outcomes. Succinylacetone provides a sensitive and specific marker for the detection of TT1 but is not universally utilized in screening protocols for the disease. Here, we report an infant transferred to our facility for evaluation and management of hyperinsulinism who subsequently developed acute-onset liver, respiratory, and renal failure around one month of life. She was found to have TT1 caused by novel pathogenic variant in fumarylacetoacetate hydrolase (c.1014 delC, p.Cys 338 Ter). Her NBS, which utilized tyrosine as a primary marker, had been reported as normal, with a tyrosine level of 151 μmol/L (reference: < 280 μmol/L). Retrospective analysis of dried blood spot samples via tandem mass spectrometry showed detectable succinylacetone ranging 4.65-10.34 μmol/L. To our knowledge, this is the first patient with TT1 whose initial presenting symptom was hyperinsulinemic hypoglycemia. The case highlights the importance of maintaining a high suspicion for metabolic disease in critically ill children, despite normal NBS. We also use the case to advocate for NBS for TT1 using succinylacetone quantitation.
Collapse
Affiliation(s)
- Jessica R. C. Priestley
- Department of Pediatrics, Division of Human Genetics, Section of Biochemical Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA; (H.A.); (I.P.-W.); (L.S.); (C.F.); (R.D.G.); (R.C.A.-N.)
- Department of Pediatrics, Pediatric Residency Program, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | - Hana Alharbi
- Department of Pediatrics, Division of Human Genetics, Section of Biochemical Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA; (H.A.); (I.P.-W.); (L.S.); (C.F.); (R.D.G.); (R.C.A.-N.)
| | - Katharine Press Callahan
- Department of Pediatrics, Division of Neonatology, Children’s Hospital of Philadelphia, PA 19104, USA;
| | - Herodes Guzman
- Department of Pediatrics, Pediatric Residency Program, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | - Irma Payan-Walters
- Department of Pediatrics, Division of Human Genetics, Section of Biochemical Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA; (H.A.); (I.P.-W.); (L.S.); (C.F.); (R.D.G.); (R.C.A.-N.)
| | - Ligia Smith
- Department of Pediatrics, Division of Human Genetics, Section of Biochemical Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA; (H.A.); (I.P.-W.); (L.S.); (C.F.); (R.D.G.); (R.C.A.-N.)
| | - Can Ficicioglu
- Department of Pediatrics, Division of Human Genetics, Section of Biochemical Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA; (H.A.); (I.P.-W.); (L.S.); (C.F.); (R.D.G.); (R.C.A.-N.)
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Rebecca D. Ganetzky
- Department of Pediatrics, Division of Human Genetics, Section of Biochemical Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA; (H.A.); (I.P.-W.); (L.S.); (C.F.); (R.D.G.); (R.C.A.-N.)
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Rebecca C. Ahrens-Nicklas
- Department of Pediatrics, Division of Human Genetics, Section of Biochemical Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA; (H.A.); (I.P.-W.); (L.S.); (C.F.); (R.D.G.); (R.C.A.-N.)
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
31
|
Singh S, Mall RV, Chaturvedi P, Yadav RR. Hereditary Tyrosinemia Type 1—A Rare Disease with Typical Radiological Features: Case Report and Review of Literature. JOURNAL OF GASTROINTESTINAL AND ABDOMINAL RADIOLOGY 2020. [DOI: 10.1055/s-0040-1708024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
AbstractHereditary tyrosinemia type 1 is one of the many inborn errors of metabolism associated with tyrosine catabolism. It is a rare disease with its incidence or prevalence in India unknown. The cascading alternate metabolism results in characteristic injury patterns to the liver, kidneys, and the central nervous system, with resultant classical radiological findings. Identifying this constellation of imaging features by a radiologist may help in arriving at a diagnosis and help referring physicians in performing appropriate biochemical tests and instituting early treatment. These usually present with chronic liver failure and are extensively evaluated for other causes of liver failure. We present a case type 1 hereditary tyrosinemia with characteristic radiological features and highlight its pathophysiology to understand the basis of these imaging findings.
Collapse
Affiliation(s)
- Somesh Singh
- Department of Radiodiagnosis, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Rana Vishwadeep Mall
- Department of Radiodiagnosis, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Pragya Chaturvedi
- Department of Radiodiagnosis, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Rajnikant R. Yadav
- Department of Radiodiagnosis, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| |
Collapse
|
32
|
Cano A, Resseguier N, Ouattara A, De Lonlay P, Arnoux JB, Brassier A, Schiff M, Pichard S, Fabre A, Hoebeke C, Guffon N, Fouilhoux A, Broué P, Touati G, Dobbelaere D, Mention K, Labarthe F, Tardieu M, De Parscau L, Feillet F, Bonnemains C, Kuster A, Labrune P, Barth M, Damaj L, Lamireau D, Berbis J, Chabrol B, Auquier P. Health Status of French Young Patients with Inborn Errors of Metabolism with Lifelong Restricted Diet. J Pediatr 2020; 220:184-192.e6. [PMID: 32145964 DOI: 10.1016/j.jpeds.2020.01.059] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/26/2019] [Accepted: 01/28/2020] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To describe the health status of young patients affected by inborn errors of metabolism that require adherence to a restricted diet (IEMRDs) and to describe and compare their self- and proxy (parent)-reported quality of life (QoL) with reference values. STUDY DESIGN A cross-sectional study was conducted in 2015-2017 in patients affected by IEMRDs (except phenylketonuria) younger than 18 years. Data collection was based on medical records, clinical examinations, parents' and children's interviews, and self-reported questionnaires. Measurements included clinical and healthcare data, child and family environment data, and self- and proxy (parent)-reported QoL. RESULTS Of the 633 eligible participants, 578 were recruited (50.3% boys; mean age: 8.7 years); their anthropometric status did not differ from the general population. Approximately one-half of them had at least 1 complication of the disease. Their self-reported global QoL did not differ from that of the general population. However, relations with friends and leisure activities QoL domains were negatively impacted, whereas relations with medical staff, relations with parents, and self-esteem QoL domains were positively impacted. Their proxy (parent)-reported QoL was negatively impacted. CONCLUSIONS Young patients affected by IEMRDs present a high rate of clinical complications. Although their proxy (parent)-reported QoL was negatively impacted, their self-reported QoL was variably impacted (both positively and negatively). These results may inform counseling for those who care for affected patients and their families.
Collapse
Affiliation(s)
- Aline Cano
- Department of Neuropediatrics and Metabolism, Reference Center of Inherited Metabolic Disorders, Timone Enfants Hospital, Marseille, France
| | - Noemie Resseguier
- Department of Epidemiology and Health Economics, AP-HM/EA 3279 CEReSS (Centre d'Etude et de Recherche sur les Services de Santé et la Qualité de vie), Aix-Marseille Univ, Marseille, France.
| | - Abdoulaye Ouattara
- Department of Epidemiology and Health Economics, AP-HM/EA 3279 CEReSS (Centre d'Etude et de Recherche sur les Services de Santé et la Qualité de vie), Aix-Marseille Univ, Marseille, France
| | - Pascale De Lonlay
- Department of Pediatric Metabolism, Reference Center of Inherited Metabolic Disorders, Necker Hospital, Paris, France
| | - Jean-Baptiste Arnoux
- Department of Pediatric Metabolism, Reference Center of Inherited Metabolic Disorders, Necker Hospital, Paris, France
| | - Anais Brassier
- Department of Pediatric Metabolism, Reference Center of Inherited Metabolic Disorders, Necker Hospital, Paris, France
| | - Manuel Schiff
- Department of Neuropediatrics and Metabolism, Reference Center of Inherited Metabolic Disorders, Robert Debré Hospital, Paris, France
| | - Samia Pichard
- Department of Neuropediatrics and Metabolism, Reference Center of Inherited Metabolic Disorders, Robert Debré Hospital, Paris, France
| | - Alexandre Fabre
- Department of Neuropediatrics and Metabolism, Reference Center of Inherited Metabolic Disorders, Timone Enfants Hospital, Marseille, France
| | - Celia Hoebeke
- Department of Neuropediatrics and Metabolism, Reference Center of Inherited Metabolic Disorders, Timone Enfants Hospital, Marseille, France
| | - Nathalie Guffon
- Department of Pediatric Metabolism, Reference Center of Inherited Metabolic Disorders, Femme Mère Enfant Hospital, Lyon, France
| | - Alain Fouilhoux
- Department of Pediatric Metabolism, Reference Center of Inherited Metabolic Disorders, Femme Mère Enfant Hospital, Lyon, France
| | - Pierre Broué
- Department of Pediatric Hepatology and Metabolism, Reference Center of Inherited Metabolic Disorders, Purpan Hospital, Toulouse, France
| | - Guy Touati
- Department of Pediatric Hepatology and Metabolism, Reference Center of Inherited Metabolic Disorders, Purpan Hospital, Toulouse, France
| | - Dries Dobbelaere
- Department of Pediatric Metabolism, Reference Center of Inherited Metabolic Disorders, Jeanne de Flandre Hospital, Lille, France
| | - Karine Mention
- Department of Pediatric Metabolism, Reference Center of Inherited Metabolic Disorders, Jeanne de Flandre Hospital, Lille, France
| | - Francois Labarthe
- Department of Pediatrics, Reference Center of Inherited Metabolic Disorders, Clocheville Hospital, Tours, France
| | - Marine Tardieu
- Department of Pediatrics, Reference Center of Inherited Metabolic Disorders, Clocheville Hospital, Tours, France
| | - Loïc De Parscau
- Department of Pediatrics, Competence Center of Inherited Metabolic Disorders, Brest Hospital, Brest, France
| | - Francois Feillet
- Department of Pediatrics, Reference Center of Inherited Metabolic Disorders, Brabois Hospital, Nancy, France
| | - Chrystèle Bonnemains
- Department of Pediatrics, Reference Center of Inherited Metabolic Disorders, Brabois Hospital, Nancy, France
| | - Alice Kuster
- Department of Pediatric Reanimation, Competence Center of Inherited Metabolic Disorders, Nantes Hospital, Nantes, France
| | - Philippe Labrune
- Department of Pediatrics, Reference Center of Rare Liver Disease, Antoine Beclere Hospital, Clamart, France
| | - Magalie Barth
- Department of Pediatrics, Competence Center of Inherited Metabolic Disorders, Angers Hospital, Angers, France
| | - Lena Damaj
- Department of Pediatrics, Competence Center of Inherited Metabolic Disorders, Rennes Hospital, Rennes, France
| | - Delphine Lamireau
- Department of Pediatrics, Competence Center of Inherited Metabolic Disorders, Pellegrin Hospital, Bordeaux, France
| | - Julie Berbis
- Department of Epidemiology and Health Economics, AP-HM/EA 3279 CEReSS (Centre d'Etude et de Recherche sur les Services de Santé et la Qualité de vie), Aix-Marseille Univ, Marseille, France
| | - Brigitte Chabrol
- Department of Neuropediatrics and Metabolism, Reference Center of Inherited Metabolic Disorders, Timone Enfants Hospital, Marseille, France
| | - Pascal Auquier
- Department of Epidemiology and Health Economics, AP-HM/EA 3279 CEReSS (Centre d'Etude et de Recherche sur les Services de Santé et la Qualité de vie), Aix-Marseille Univ, Marseille, France
| |
Collapse
|
33
|
Yilmaz O, Daly A, Pinto A, Ashmore C, Evans S, Gupte G, Santra S, Preece MA, Mckiernan P, Kitchen S, Yabanci Ayhan N, MacDonald A. Natural Protein Tolerance and Metabolic Control in Patients with Hereditary Tyrosinaemia Type 1. Nutrients 2020; 12:E1148. [PMID: 32325917 PMCID: PMC7230348 DOI: 10.3390/nu12041148] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/15/2020] [Accepted: 04/15/2020] [Indexed: 12/23/2022] Open
Abstract
In a longitudinal retrospective study, we aimed to assess natural protein (NP) tolerance and metabolic control in a cohort of 20 Hereditary Tyrosinaemia type I (HTI) patients. Their median age was 12 years ([3.2-17.7 years], n = 11 female, n = 8 Caucasian, n = 8 Asian origin, n = 2 Arabic and n = 2 Indian). All were on nitisinone (NTBC) with a median dose of 0.7 g/kg/day (range 0.4-1.5 g/kg/day) and were prescribed a tyrosine (Tyr)/phenylalanine (Phe)-restricted diet supplemented with Tyr/Phe-free L-amino acids. Data were collected on clinical signs at presentation, medical history, annual dietary prescriptions, and blood Phe and Tyr levels from diagnosis until transition to the adult service (aged 16-18 years) or liver transplantation (if it preceded transition). The median age of diagnosis was 2 months (range: 0 to 24 months), with n = 1 diagnosed by newborn screening, n = 3 following phenylketonuria (PKU) screening and n = 7 by sibling screening. Five patients were transplanted (median age 6.3 years), and one died due to liver cancer. The median follow-up was 10 years (3-16 years), and daily prescribed NP intake increased from a median of 5 to 24 g/day. Lifetime median blood Tyr (370 µmol/L, range 280-420 µmol/L) and Phe (50 µmol/L, 45-70 µmol/L) were maintained within the target recommended ranges. This cohort of HTI patients were able to increase the daily NP intake with age while maintaining good metabolic control. Extra NP may improve lifelong adherence to the diet.
Collapse
Affiliation(s)
- Ozlem Yilmaz
- Birmingham Women’s and Children’s Hospital, Birmingham B4 6NH, UK; (O.Y.); (A.D.); (A.P.); (C.A.); (S.E.); (G.G.); (S.S.); (M.A.P.); (S.K.)
- Department of Nutrition and Dietetics, Ankara Yildirim Beyazit University, 06760 Ankara, Turkey
| | - Anne Daly
- Birmingham Women’s and Children’s Hospital, Birmingham B4 6NH, UK; (O.Y.); (A.D.); (A.P.); (C.A.); (S.E.); (G.G.); (S.S.); (M.A.P.); (S.K.)
| | - Alex Pinto
- Birmingham Women’s and Children’s Hospital, Birmingham B4 6NH, UK; (O.Y.); (A.D.); (A.P.); (C.A.); (S.E.); (G.G.); (S.S.); (M.A.P.); (S.K.)
| | - Catherine Ashmore
- Birmingham Women’s and Children’s Hospital, Birmingham B4 6NH, UK; (O.Y.); (A.D.); (A.P.); (C.A.); (S.E.); (G.G.); (S.S.); (M.A.P.); (S.K.)
| | - Sharon Evans
- Birmingham Women’s and Children’s Hospital, Birmingham B4 6NH, UK; (O.Y.); (A.D.); (A.P.); (C.A.); (S.E.); (G.G.); (S.S.); (M.A.P.); (S.K.)
| | - Girish Gupte
- Birmingham Women’s and Children’s Hospital, Birmingham B4 6NH, UK; (O.Y.); (A.D.); (A.P.); (C.A.); (S.E.); (G.G.); (S.S.); (M.A.P.); (S.K.)
| | - Saikat Santra
- Birmingham Women’s and Children’s Hospital, Birmingham B4 6NH, UK; (O.Y.); (A.D.); (A.P.); (C.A.); (S.E.); (G.G.); (S.S.); (M.A.P.); (S.K.)
| | - Mary Anne Preece
- Birmingham Women’s and Children’s Hospital, Birmingham B4 6NH, UK; (O.Y.); (A.D.); (A.P.); (C.A.); (S.E.); (G.G.); (S.S.); (M.A.P.); (S.K.)
| | - Patrick Mckiernan
- Gastroenterology/ Hepatic/Nutrition, UPMC, Children’s Hospital of Pittsburg, Pittsburg, PA 15224, USA;
| | - Steve Kitchen
- Birmingham Women’s and Children’s Hospital, Birmingham B4 6NH, UK; (O.Y.); (A.D.); (A.P.); (C.A.); (S.E.); (G.G.); (S.S.); (M.A.P.); (S.K.)
| | | | - Anita MacDonald
- Birmingham Women’s and Children’s Hospital, Birmingham B4 6NH, UK; (O.Y.); (A.D.); (A.P.); (C.A.); (S.E.); (G.G.); (S.S.); (M.A.P.); (S.K.)
| |
Collapse
|
34
|
Combined 3D-quantitative structure-activity relationships and topomer technology-based molecular design of human 4-hydroxyphenylpyruvate dioxygenase inhibitors. Future Med Chem 2020; 12:795-811. [PMID: 32223563 DOI: 10.4155/fmc-2019-0349] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: 4-Hydroxyphenylpyruvate dioxygenase (HPPD) has attracted increasing attention as an important target against tyrosinemia type I. This paper aimed to explore the structure-activity relationship of HPPD inhibitors with pyrazole scaffolds and to design novel HPPD inhibitors. Methodology & results: The best 3D-quantitative structure-activity relationships model was established by two different strategies based on 40 pyrazole scaffold-based analogs. Screening of molecular fragments by topomer technology, combined with molecular docking, 14 structures were identified for potential human HPPD inhibitory activity. Molecular dynamics results demonstrated that all the compounds obtained bound to the enzyme and possessed a satisfactory binding free energy. Conclusion: The quantitative structure-activity relationship of HPPD inhibitors of pyrazole scaffolds was clarified and 14 original structures with potential human HPPD inhibitory activity were obtained.
Collapse
|
35
|
Barone H, Bliksrud YT, Elgen IB, Szigetvari PD, Kleppe R, Ghorbani S, Hansen EV, Haavik J. Tyrosinemia Type 1 and symptoms of ADHD: Biochemical mechanisms and implications for treatment and prognosis. Am J Med Genet B Neuropsychiatr Genet 2020; 183:95-105. [PMID: 31633311 DOI: 10.1002/ajmg.b.32764] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/25/2019] [Accepted: 09/17/2019] [Indexed: 12/20/2022]
Abstract
Hereditary tyrosinemia Type 1 (HT-1) is a rare metabolic disease where the enzyme catalyzing the final step of tyrosine breakdown is defect, leading to accumulation of toxic metabolites. Nitisinone inhibits the degradation of tyrosine and thereby the production of harmful metabolites, however, the concentration of tyrosine also increases. We investigated the relationship between plasma tyrosine concentrations and cognitive functions and how tyrosine levels affected enzyme activities of human tyrosine hydroxylase (TH) and tryptophan hydroxylase 2 (TPH2). Eight Norwegian children between 6 and 18 years with HT-1 were assessed using questionnaires measuring Attention Deficit Hyperactivity Disorder (ADHD)-symptoms and executive functioning. Recent and past levels of tyrosine were measured and the enzyme activities of TH and TPH2 were studied at conditions replicating normal and pathological tyrosine concentrations. We observed a significant positive correlation between mean tyrosine levels and inattention symptoms. While TH exhibited prominent substrate inhibition kinetics, TPH2 activity also decreased at elevated tyrosine levels. Inhibition of both enzymes may impair syntheses of dopamine, noradrenaline, and serotonin in brain tissue. Inattention in treated HT-1 patients may be related to decreased production of these monoamines. Our results support recommendations of strict guidelines on plasma tyrosine levels in HT-1. ADHD-related deficits, particularly inattention, should be monitored in HT-1 patients to determine whether intervention is necessary.
Collapse
Affiliation(s)
- Helene Barone
- Department of Child and Adolescent Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Yngve T Bliksrud
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Irene B Elgen
- Department of Child and Adolescent Psychiatry, Haukeland University Hospital, Bergen, Norway
| | | | - Rune Kleppe
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Sadaf Ghorbani
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Eirik V Hansen
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Jan Haavik
- Department of Biomedicine, University of Bergen, Bergen, Norway.,Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
36
|
Hughes JH, Wilson PJM, Sutherland H, Judd S, Hughes AT, Milan AM, Jarvis JC, Bou‐Gharios G, Ranganath LR, Gallagher JA. Dietary restriction of tyrosine and phenylalanine lowers tyrosinemia associated with nitisinone therapy of alkaptonuria. J Inherit Metab Dis 2020; 43:259-268. [PMID: 31503358 PMCID: PMC7079096 DOI: 10.1002/jimd.12172] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/03/2019] [Accepted: 09/09/2019] [Indexed: 12/12/2022]
Abstract
Alkaptonuria (AKU) is caused by homogentisate 1,2-dioxygenase deficiency that leads to homogentisic acid (HGA) accumulation, ochronosis and severe osteoarthropathy. Recently, nitisinone treatment, which blocks HGA formation, has been effective in AKU patients. However, a consequence of nitisinone is elevated tyrosine that can cause keratopathy. The effect of tyrosine and phenylalanine dietary restriction was investigated in nitisinone-treated AKU mice, and in an observational study of dietary intervention in AKU patients. Nitisinone-treated AKU mice were fed tyrosine/phenylalanine-free and phenylalanine-free diets with phenylalanine supplementation in drinking water. Tyrosine metabolites were measured pre-nitisinone, post-nitisinone, and after dietary restriction. Subsequently an observational study was undertaken in 10 patients attending the National Alkaptonuria Centre (NAC), with tyrosine >700 μmol/L who had been advised to restrict dietary protein intake and where necessary, to use tyrosine/phenylalanine-free amino acid supplements. Elevated tyrosine (813 μmol/L) was significantly reduced in nitisinone-treated AKU mice fed a tyrosine/phenylalanine-free diet in a dose responsive manner. At 3 days of restriction, tyrosine was 389.3, 274.8, and 144.3 μmol/L with decreasing phenylalanine doses. In contrast, tyrosine was not effectively reduced in mice by a phenylalanine-free diet; at 3 days tyrosine was 757.3, 530.2, and 656.2 μmol/L, with no dose response to phenylalanine supplementation. In NAC patients, tyrosine was significantly reduced (P = .002) when restricting dietary protein alone, and when combined with tyrosine/phenylalanine-free amino acid supplementation; 4 out of 10 patients achieved tyrosine <700 μmol/L. Tyrosine/phenylalanine dietary restriction significantly reduced nitisinone-induced tyrosinemia in mice, with phenylalanine restriction alone proving ineffective. Similarly, protein restriction significantly reduced circulating tyrosine in AKU patients.
Collapse
Affiliation(s)
- Juliette H. Hughes
- Department of Musculoskeletal Biology I, Institute of Ageing and Chronic DiseaseUniversity of LiverpoolLiverpoolUK
| | - Peter J. M. Wilson
- Department of Musculoskeletal Biology I, Institute of Ageing and Chronic DiseaseUniversity of LiverpoolLiverpoolUK
| | - Hazel Sutherland
- Department of Musculoskeletal Biology I, Institute of Ageing and Chronic DiseaseUniversity of LiverpoolLiverpoolUK
| | - Shirley Judd
- Department of Nutrition and DieteticsRoyal Liverpool University Hospital TrustLiverpoolUK
| | - Andrew T. Hughes
- Department of Musculoskeletal Biology I, Institute of Ageing and Chronic DiseaseUniversity of LiverpoolLiverpoolUK
- Liverpool Clinical Laboratories, Department of Clinical Biochemistry and Metabolic MedicineRoyal Liverpool and Broadgreen University Hospitals TrustLiverpoolUK
| | - Anna M. Milan
- Department of Musculoskeletal Biology I, Institute of Ageing and Chronic DiseaseUniversity of LiverpoolLiverpoolUK
- Liverpool Clinical Laboratories, Department of Clinical Biochemistry and Metabolic MedicineRoyal Liverpool and Broadgreen University Hospitals TrustLiverpoolUK
| | - Jonathan C. Jarvis
- School of Sport and Exercise Sciences, Faculty of ScienceLiverpool John Moores UniversityLiverpoolUK
| | - George Bou‐Gharios
- Department of Musculoskeletal Biology I, Institute of Ageing and Chronic DiseaseUniversity of LiverpoolLiverpoolUK
| | - Lakshminarayan R. Ranganath
- Department of Musculoskeletal Biology I, Institute of Ageing and Chronic DiseaseUniversity of LiverpoolLiverpoolUK
- Liverpool Clinical Laboratories, Department of Clinical Biochemistry and Metabolic MedicineRoyal Liverpool and Broadgreen University Hospitals TrustLiverpoolUK
| | - James A. Gallagher
- Department of Musculoskeletal Biology I, Institute of Ageing and Chronic DiseaseUniversity of LiverpoolLiverpoolUK
| |
Collapse
|
37
|
Liu Y, Luo Y, Xia L, Qiu BJ, Zhou T, Feng MX, Xue F, Chen XS, Han LS, Zhang JJ, Xia Q. Living-donor liver transplantation for children with tyrosinemia type I. J Dig Dis 2020; 21:189-194. [PMID: 31953907 DOI: 10.1111/1751-2980.12846] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To evaluate the efficacy of living-donor liver transplantation (LDLT) in children with tyrosinemia type I. METHODS Altogether 10 patients diagnosed with tyrosinemia type I underwent LDLT between June 2013 and April 2019. Cirrhosis was the indication for LDLT in all 10 patients, and hepatocellular carcinoma (HCC) was suspected in nine. Patients' outcomes, including liver function, restoration of metabolism, quality of life and physical development, were analyzed after LDLT. RESULTS All recipients were alive with a normal liver function after a median follow-up period of 49 months. Pathological examinations detected HCC in one patient, dysplasia in five and cirrhosis in all. Nine patients were found to have elevated alpha-fetoprotein level, and their median alpha-fetoprotein level dropped from 2520 ng/mL to a normal level after LDLT, with no recurrence of HCC detected during the follow-up. Tyrosine metabolism was restored to its normal level with normalized plasma tyrosine and succinylacetone concentrations. Moreover, urinary succinylacetone excretion decreased significantly during the follow up. LDLT improved patients' renal tubular function, as evidenced by the normalized plasma phosphate concentration and improved glomerular filtration rate. Severe rickets symptoms, including spontaneous fractures and bone pain, were also ameliorated. Improved motor function was reported by all patients' parents during the follow-up. Dietary restriction was no longer required, which was associated with a favorable catch-up in growth and improved quality of life. Complete resolution of hypertrophic cardiomyopathy was observed one year after LDLT in one patient. CONCLUSION LDLT is an effective treatment for patients with end-stage liver disease resulting from tyrosinemia type I.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Luo
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bi Jun Qiu
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Zhou
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ming Xuan Feng
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Xue
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao Song Chen
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lian Shu Han
- Department of Pediatrics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Jun Zhang
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
38
|
van Vliet K, van Ginkel WG, Jahja R, Daly A, MacDonald A, De Laet C, Vara R, Rahman Y, Cassiman D, Eyskens F, Timmer C, Mumford N, Bierau J, van Hasselt PM, Gissen P, Goyens PJ, McKiernan PJ, Wilcox G, Morris AAM, Jameson EA, Huijbregts SCJ, van Spronsen FJ. Emotional and behavioral problems, quality of life and metabolic control in NTBC-treated Tyrosinemia type 1 patients. Orphanet J Rare Dis 2019; 14:285. [PMID: 31801588 PMCID: PMC6894144 DOI: 10.1186/s13023-019-1259-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/22/2019] [Indexed: 02/06/2023] Open
Abstract
Abstract Background Treatment with 2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione (NTBC) and dietary phenylalanine and tyrosine restriction improves physical health and life expectancy in Tyrosinemia type 1 (TT1). However, neurocognitive outcome is suboptimal. This study aimed to investigate behavior problems and health-related quality of life (HR-QoL) in NTBC-dietary-treated TT1 and to relate this to phenylalanine and tyrosine concentrations. Results Thirty-one TT1 patients (19 males; mean age 13.9 ± 5.3 years) were included in this study. Emotional and behavioral problems, as measured by the Achenbach System of Empirically Based Assessment, were present in almost all domains. Attention and thought problems were particularly evident. HR-QoL was assessed by the TNO AZL Children’s and Adults QoL questionnaires. Poorer HR-QoL as compared to reference populations was observed for the domains: independent daily functioning, cognitive functioning and school performance, social contacts, motor functioning, and vitality. Both internalizing and externalizing behavior problems were associated with low phenylalanine (and associated lower tyrosine) concentrations during the first year of life. In contrast, high tyrosine (and associated higher phenylalanine) concentrations during life and specifically the last year before testing were associated with more internalizing behavior and/or HR-QoL problems. Conclusions TT1 patients showed several behavior problems and a lower HR-QoL. Associations with metabolic control differed for different age periods. This suggests the need for continuous fine-tuning and monitoring of dietary treatment to keep phenylalanine and tyrosine concentrations within target ranges in NTBC-treated TT1 patients.
Collapse
Affiliation(s)
- Kimber van Vliet
- Beatrix Children's Hospital, Groningen, Division of Metabolic Diseases, University of Groningen, University Medical Center Groningen, CA33, PO box 30.001, 9700 RB, Groningen, Netherlands
| | - Willem G van Ginkel
- Beatrix Children's Hospital, Groningen, Division of Metabolic Diseases, University of Groningen, University Medical Center Groningen, CA33, PO box 30.001, 9700 RB, Groningen, Netherlands
| | - Rianne Jahja
- Beatrix Children's Hospital, Groningen, Division of Metabolic Diseases, University of Groningen, University Medical Center Groningen, CA33, PO box 30.001, 9700 RB, Groningen, Netherlands
| | - Anne Daly
- Birmingham Children's Hospital, Birmingham, UK
| | | | - Corinne De Laet
- Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles, Brussels, Belgium
| | - Roshni Vara
- Evelina London Children's Hospital, London, UK
| | | | - David Cassiman
- University Hospital Gasthuisberg, University of Leuven, Leuven, Belgium
| | - Francois Eyskens
- Kon. Mathilde Moeder- en Kindcentrum, University Hospital of Antwerp, Antwerp, Belgium
| | | | - Nicky Mumford
- The NIHR Great Ormond Street Hospital Biomedical Research Centre (BRC ), London, UK
| | - Jörgen Bierau
- Maastricht University Medical Center, Maastricht, Netherlands
| | - Peter M van Hasselt
- Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Utrecht, Netherlands
| | - Paul Gissen
- The NIHR Great Ormond Street Hospital Biomedical Research Centre (BRC ), London, UK
| | - Philippe J Goyens
- Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Gisela Wilcox
- School of Medical Sciences, Faculty of Biology Medicine & Health, University of Manchester, Manchester, UK.,The Mark Holland Metabolic Unit, Salford Royal Foundation NHS Trust, Greater Manchester, M6 8HD, Salford, UK
| | - Andrew A M Morris
- Willink Metabolic Unit, Manchester Centre for Genomic Medicine, Manchester University Hospitals NHS Foundation Trust, St Mary's Hospital, Manchester, UK
| | - Elisabeth A Jameson
- Willink Metabolic Unit, Manchester Centre for Genomic Medicine, Manchester University Hospitals NHS Foundation Trust, St Mary's Hospital, Manchester, UK
| | - Stephan C J Huijbregts
- University of Leiden, Clinical Child and Adolescent Studies: Neurodevelopmental Disorders, Leiden, Netherlands
| | - Francjan J van Spronsen
- Beatrix Children's Hospital, Groningen, Division of Metabolic Diseases, University of Groningen, University Medical Center Groningen, CA33, PO box 30.001, 9700 RB, Groningen, Netherlands.
| |
Collapse
|
39
|
van Ginkel WG, Rodenburg IL, Harding CO, Hollak CEM, Heiner-Fokkema MR, van Spronsen FJ. Long-Term Outcomes and Practical Considerations in the Pharmacological Management of Tyrosinemia Type 1. Paediatr Drugs 2019; 21:413-426. [PMID: 31667718 PMCID: PMC6885500 DOI: 10.1007/s40272-019-00364-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tyrosinemia type 1 (TT1) is a rare metabolic disease caused by a defect in tyrosine catabolism. TT1 is clinically characterized by acute liver failure, development of hepatocellular carcinoma, renal and neurological problems, and consequently an extremely poor outcome. This review showed that the introduction of 2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione (NTBC) in 1992 has revolutionized the outcome of TT1 patients, especially when started pre-clinically. If started early, NTBC can prevent liver failure, renal problems, and neurological attacks and decrease the risk for hepatocellular carcinoma. NTBC has been shown to be safe and well tolerated, although the long-term effectiveness of treatment with NTBC needs to be awaited. The high tyrosine concentrations caused by treatment with NTBC could result in ophthalmological and skin problems and requires life-long dietary restriction of tyrosine and its precursor phenylalanine, which could be strenuous to adhere to. In addition, neurocognitive problems have been reported since the introduction of NTBC, with hypothesized but as yet unproven pathophysiological mechanisms. Further research should be done to investigate the possible relationship between important clinical outcomes and blood concentrations of biochemical parameters such as phenylalanine, tyrosine, succinylacetone, and NTBC, and to develop clear guidelines for treatment and follow-up with reliable measurements. This all in order to ultimately improve the combined NTBC and dietary treatment and limit possible complications such as hepatocellular carcinoma development, neurocognitive problems, and impaired quality of life.
Collapse
Affiliation(s)
- Willem G van Ginkel
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700 RB, Groningen, The Netherlands
| | - Iris L Rodenburg
- Department of Dietetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Cary O Harding
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, USA
| | - Carla E M Hollak
- Deparment of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - M Rebecca Heiner-Fokkema
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Francjan J van Spronsen
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700 RB, Groningen, The Netherlands.
| |
Collapse
|
40
|
Blood and Brain Biochemistry and Behaviour in NTBC and Dietary Treated Tyrosinemia Type 1 Mice. Nutrients 2019; 11:nu11102486. [PMID: 31623189 PMCID: PMC6836052 DOI: 10.3390/nu11102486] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 10/11/2019] [Accepted: 10/14/2019] [Indexed: 02/05/2023] Open
Abstract
Tyrosinemia type 1 (TT1) is a rare metabolic disease caused by a defect in the tyrosine degradation pathway. Neurocognitive deficiencies have been described in TT1 patients, that have, among others, been related to changes in plasma large neutral amino acids (LNAA) that could result in changes in brain LNAA and neurotransmitter concentrations. Therefore, this project aimed to investigate plasma and brain LNAA, brain neurotransmitter concentrations and behavior in C57 Bl/6 fumarylacetoacetate hydrolase deficient (FAH−/−) mice treated with 2-(2-nitro-4-trifluoromethylbenoyl)-1,3-cyclohexanedione (NTBC) and/or diet and wild-type mice. Plasma and brain tyrosine concentrations were clearly increased in all NTBC treated animals, even with diet (p < 0.001). Plasma and brain phenylalanine concentrations tended to be lower in all FAH−/− mice. Other brain LNAA, were often slightly lower in NTBC treated FAH−/− mice. Brain neurotransmitter concentrations were usually within a normal range, although serotonin was negatively correlated with brain tyrosine concentrations (p < 0.001). No clear behavioral differences between the different groups of mice could be found. To conclude, this is the first study measuring plasma and brain biochemistry in FAH−/− mice. Clear changes in plasma and brain LNAA have been shown. Further research should be done to relate the biochemical changes to neurocognitive impairments in TT1 patients.
Collapse
|
41
|
Taylor AM, Shepherd L. The potential of nitisinone for the treatment of alkaptonuria. Expert Opin Orphan Drugs 2019. [DOI: 10.1080/21678707.2019.1664899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Adam M Taylor
- Lancaster Medical School, Lancaster University, Lancaster, UK
| | - Laura Shepherd
- Lancaster Medical School, Lancaster University, Lancaster, UK
| |
Collapse
|
42
|
Couce ML, Sánchez-Pintos P, Aldámiz-Echevarría L, Vitoria I, Navas V, Martín-Hernández E, García-Volpe C, Pintos G, Peña-Quintana L, Hernández T, Gil D, Sánchez-Valverde F, Bueno M, Roca I, López-Ruzafa E, Díaz-Fernández C. Evolution of tyrosinemia type 1 disease in patients treated with nitisinone in Spain. Medicine (Baltimore) 2019; 98:e17303. [PMID: 31574857 PMCID: PMC6775438 DOI: 10.1097/md.0000000000017303] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 07/19/2019] [Accepted: 08/27/2019] [Indexed: 01/01/2023] Open
Abstract
Treatment with nitisinone (NTBC) has brought about a drastic improvement in the treatment and prognosis of hereditary tyrosinemia type I (HT1). We conducted a retrospective observational multicentric study in Spanish HT1 patients treated with NTBC to assess clinical and biochemical long-term evolution.We evaluated 52 patients, 7 adults and 45 children, treated with NTBC considering: age at diagnosis, diagnosis by clinical symptoms, or by newborn screening (NBS); phenotype (acute/subacute/chronic), mutational analysis; symptoms at diagnosis and clinical course; biochemical markers; doses of NTBC; treatment adherence; anthropometric evolution; and neurocognitive outcome.The average follow-up period was 6.1 ± 4.9 and 10.6 ± 5.4 years in patients with early and late diagnosis respectively. All patients received NTBC from diagnosis with an average dose of 0.82 mg/kg/d. All NBS-patients (n = 8) were asymptomatic at diagnosis except 1 case with acute liver failure, and all remain free of liver and renal disease in follow-up. Liver and renal affectation was markedly more frequent at diagnosis in patients with late diagnosis (P < .001 and .03, respectively), with ulterior positive hepatic and renal course in 86.4% and 93.2% of no-NBS patients, although 1 patient with good metabolic control developed hepatocarcinoma.Despite a satisfactory global nutritional evolution, 46.1% of patients showed overweight/obesity. Interestingly lower body mass index was observed in patients with good dietary adherence (20.40 ± 4.43 vs 24.30 ± 6.10; P = .08) and those with good pharmacological adherence (21.19 ± 4.68 vs 28.58 ± 213.79).intellectual quotient was ≥85 in all NBS- and 68.75% of late diagnosis cases evaluated, 15% of which need pedagogical support, and 6.8% (3/44) showed school failure.Among the 12 variants identified in fumarylacetoacetate hydrolase gene, 1 of them novel (H63D), the most prevalent in Spanish population is c.554-1 G>T.After NTBC treatment a reduction in tyrosine and alpha-fetoprotein levels was observed in all the study groups, significant for alpha-fetoprotein in no NBS-group (P = .03), especially in subacute/chronic forms (P = .018).This series confirms that NTBC treatment had clearly improved the prognosis and quality of life of HT1 patients, but it also shows frequent cognitive dysfunctions and learning difficulties in medium-term follow-up, and, in a novel way, a high percentage of overweight/obesity.
Collapse
Affiliation(s)
- María Luz Couce
- Unit of Diagnosis and Treatment of Congenital Metabolic Diseases, S. Neonatology, Department of Pediatrics, Clínico Universitario de Santiago de Compostela, CIBERER, Health Research Institute of Santiago de Compostela (IDIS)
| | - Paula Sánchez-Pintos
- Unit of Diagnosis and Treatment of Congenital Metabolic Diseases, S. Neonatology, Department of Pediatrics, Clínico Universitario de Santiago de Compostela, CIBERER, Health Research Institute of Santiago de Compostela (IDIS)
| | - Luís Aldámiz-Echevarría
- Unit of Metabolism, Department of Pediatrics, Hospital de Cruces, Group of Metabolism, Biocruces Health Research Institute, CIBERER
| | | | - Victor Navas
- Pediatric Gastroenterology and Nutrition Unit Hospital Carlos Haya, Málaga
| | | | - Camila García-Volpe
- Pediatric Gastroenterology, Hepatology and Nutrition Unit, H. San Joan de Deu, Barcelona
| | | | - Luis Peña-Quintana
- Gastroenterology and Nutrition Unit Complejo Hospitalario Universitario Insular-Materno Infantil, CIBEROBN, Las Palmas de Gran Canaria University, Las Palmas
| | | | - David Gil
- Pediatric Gastroenterology, Hepatology and Nutrition Unit Hospital Virgen da Arrixaca, Murcia
| | | | - María Bueno
- Metabolic Congenital Diseases Unit, Hospital Virgen del Rocío, Sevilla
| | - Iria Roca
- Unit of Diagnosis and Treatment of Congenital Metabolic Diseases, S. Neonatology, Department of Pediatrics, Clínico Universitario de Santiago de Compostela, CIBERER, Health Research Institute of Santiago de Compostela (IDIS)
| | | | - Carmen Díaz-Fernández
- Unit of Hepatology and Infantile Hepatic Transplantation, Hospital Universitario La Paz, Madrid, Spain
| |
Collapse
|
43
|
Karaca CA, Yilmaz C, Farajov R, Iakobadze Z, Aydogdu S, Kilic M. Live donor liver transplantation for type 1 tyrosinemia: An analysis of 15 patients. Pediatr Transplant 2019; 23:e13498. [PMID: 31155831 DOI: 10.1111/petr.13498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/10/2019] [Accepted: 05/03/2019] [Indexed: 12/19/2022]
Abstract
Type 1 tyrosinemia is a rare metabolic disorder of the tyrosine degradation pathway. Due to the rarity of the disease, the best evidence literature offers is limited to guidelines based on expert opinions and optimal treatment is still a debate. LT serves as a definitive treatment of the defective metabolic pathway in the liver along with other serious disease manifestations such as LF and HCC. Nitisinone is a relatively new agent that is currently recommended for the medical management of the disease. Its mechanism of action is well understood, and efficacy is well established when started presymptomatically. This study aims to evaluate outcomes of 15 patients with type 1 tyrosinemia who underwent LT in nitisinone era and discuss its effect on prevention of HCC. A LT database of 1037 patients was reviewed. Data from 15 patients with type 1 tyrosinemia were retrospectively analyzed. All the patients except one were treated with nitisinone prior to LT. Most common indications for LT were LF and suspicious nodules. Seven patients had HCC. Mortality rate was 20% (n = 3). Nitisinone treatment has opened new horizons in the management of type 1 tyrosinemia, but LT still remains the only option for the patients developing LF and in the event of HCC. Neonatal screening programs utilizing blood succinyl acetone as the marker should be encouraged especially in the countries, such as Turkey, with high prevalence of consanguineous marriages.
Collapse
Affiliation(s)
- Can A Karaca
- Faculty of Medicine, Izmir University of Economics, Izmir, Turkey
| | - Cahit Yilmaz
- Department of Liver Transplantation, Izmir Kent Hospital, Izmir, Turkey
| | - Rasim Farajov
- Department of Liver Transplantation, Izmir Kent Hospital, Izmir, Turkey
| | - Zaza Iakobadze
- Department of Liver Transplantation, Izmir Kent Hospital, Izmir, Turkey
| | - Sema Aydogdu
- Department of Pediatric Gastroenterology, Ege University Faculty of Medicine, Izmir, Turkey
| | - Murat Kilic
- Department of Liver Transplantation, Izmir Kent Hospital, Izmir, Turkey
| |
Collapse
|
44
|
Sloboda N, Wiedemann A, Merten M, Alqahtani A, Jeannesson E, Blum A, Henn-Ménétré S, Guéant JL, Renard E, Feillet F. Efficacy of low dose nitisinone in the management of alkaptonuria. Mol Genet Metab 2019; 127:184-190. [PMID: 31235217 DOI: 10.1016/j.ymgme.2019.06.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 06/17/2019] [Accepted: 06/17/2019] [Indexed: 02/07/2023]
Abstract
AIM To study the efficacy of low dosage of nitisinone in alkaptonuria. BACKGROUND Alkaptonuria (AKU) is a rare genetic disease which induces deposition of homogentisic acid (HGA) in connective inducing premature arthritis, lithiasis, cardiac valve disease, fractures, muscle and tendon ruptures and osteopenia. Recent studies showed that nitisinone decreases HGA and is a beneficial therapy in AKU. This treatment induces an increase in tyrosine levels which can induces adverse effects as keratopathy. METHODS We described the evolution HGA excretion and tyrosine evolution in 3 AKU patients treated by very low dosage of nitisinone with regards to their daily protein intakes. We also described the first pregnancy in an AKU patient treated by nitisinone. RESULTS We found mild clinical signs of alkaptonuria on vertebra MRI in two young adults and homogentisate deposition in teeth of a 5 years old girl. Very low dose of nitisinone (10% of present recommended dose: 0.2 mg/day) allowed to decrease homogentisic acid by >90% without increasing tyrosine levels above 500 μmol/ in these three patients. INTERPRETATIONS The analysis of the follow-up data shows that, in our three patients, a low-dosage of nitisinone is sufficient to decrease urinary HGA without increasing plasma tyrosine levels above the threshold of 500 μmol/L.
Collapse
Affiliation(s)
- Natacha Sloboda
- Pediatric Unit, Reference center for Inborn Errors of Metabolism of Nancy, University Children's Hospital, CHU Brabois, Vandoeuvre les Nancy, France
| | - Arnaud Wiedemann
- Pediatric Unit, Reference center for Inborn Errors of Metabolism of Nancy, University Children's Hospital, CHU Brabois, Vandoeuvre les Nancy, France; INSERM UMRS 1256 NGERE, Nutrition, Genetics, and Environmental Risk Exposure, National Center of Inborn Errors of Metabolism, Faculty of Medicine of Nancy, University of Lorraine and University Regional Hospital Center of Nancy, Vandoeuvre-les-Nancy F-54000, France
| | - Marc Merten
- INSERM UMRS 1256 NGERE, Nutrition, Genetics, and Environmental Risk Exposure, National Center of Inborn Errors of Metabolism, Faculty of Medicine of Nancy, University of Lorraine and University Regional Hospital Center of Nancy, Vandoeuvre-les-Nancy F-54000, France; Biochemistry and Molecular Biology Laboratory (Nutrition, Metabolism), CHRU de Nancy, Nancy, France
| | - Amerh Alqahtani
- Pediatric Unit, Reference center for Inborn Errors of Metabolism of Nancy, University Children's Hospital, CHU Brabois, Vandoeuvre les Nancy, France
| | - Elise Jeannesson
- INSERM UMRS 1256 NGERE, Nutrition, Genetics, and Environmental Risk Exposure, National Center of Inborn Errors of Metabolism, Faculty of Medicine of Nancy, University of Lorraine and University Regional Hospital Center of Nancy, Vandoeuvre-les-Nancy F-54000, France; Biochemistry and Molecular Biology Laboratory (Nutrition, Metabolism), CHRU de Nancy, Nancy, France
| | - Alain Blum
- Pediatric Unit, Reference center for Inborn Errors of Metabolism of Nancy, University Children's Hospital, CHU Brabois, Vandoeuvre les Nancy, France; Radiology Unit Guilloz, University Hospital, Hôpital Central, Nancy, France
| | - Sophie Henn-Ménétré
- Pediatric Unit, Reference center for Inborn Errors of Metabolism of Nancy, University Children's Hospital, CHU Brabois, Vandoeuvre les Nancy, France; Pharmacy Unit, University Hospital, CHU Brabois, Vandoeuvre les Nancy, France
| | - Jean-Louis Guéant
- INSERM UMRS 1256 NGERE, Nutrition, Genetics, and Environmental Risk Exposure, National Center of Inborn Errors of Metabolism, Faculty of Medicine of Nancy, University of Lorraine and University Regional Hospital Center of Nancy, Vandoeuvre-les-Nancy F-54000, France; Biochemistry and Molecular Biology Laboratory (Nutrition, Metabolism), CHRU de Nancy, Nancy, France
| | - Emeline Renard
- Pediatric Unit, Reference center for Inborn Errors of Metabolism of Nancy, University Children's Hospital, CHU Brabois, Vandoeuvre les Nancy, France; INSERM UMRS 1256 NGERE, Nutrition, Genetics, and Environmental Risk Exposure, National Center of Inborn Errors of Metabolism, Faculty of Medicine of Nancy, University of Lorraine and University Regional Hospital Center of Nancy, Vandoeuvre-les-Nancy F-54000, France
| | - François Feillet
- Pediatric Unit, Reference center for Inborn Errors of Metabolism of Nancy, University Children's Hospital, CHU Brabois, Vandoeuvre les Nancy, France; INSERM UMRS 1256 NGERE, Nutrition, Genetics, and Environmental Risk Exposure, National Center of Inborn Errors of Metabolism, Faculty of Medicine of Nancy, University of Lorraine and University Regional Hospital Center of Nancy, Vandoeuvre-les-Nancy F-54000, France.
| |
Collapse
|
45
|
Ozcan HN, Karcaaltincaba M, Pektas E, Sivri HS, Oguz B, Dursun A, Tokatli A, Coskun T, Haliloglu M. Imaging liver nodules in tyrosinemia type-1: A retrospective review of 16 cases in a tertiary pediatric hospital. Eur J Radiol 2019; 116:41-46. [DOI: 10.1016/j.ejrad.2019.04.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 03/07/2019] [Accepted: 04/23/2019] [Indexed: 11/28/2022]
|
46
|
Kaiser RA, Nicolas CT, Allen KL, Chilton JA, Du Z, Hickey RD, Lillegard JB. Hepatotoxicity and Toxicology of In Vivo Lentiviral Vector Administration in Healthy and Liver-Injury Mouse Models. HUM GENE THER CL DEV 2019; 30:57-66. [PMID: 30860398 PMCID: PMC6589498 DOI: 10.1089/humc.2018.249] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/07/2019] [Indexed: 12/31/2022] Open
Abstract
General safety and toxicology assessments supporting in vivo lentiviral vector-based therapeutic development are sparse. We have previously demonstrated the efficacy of a lentiviral vector expressing fumarylacetoacetate hydrolase (LV-FAH) to cure animal models of hereditary tyrosinemia type 1. Therefore, we performed a complete preclinical toxicological evaluation of LV-FAH, in a large cohort (n = 20/group) of wildtype mice and included matched groups of N-nitrosodiethylamine/carbon tetrachloride (DEN/CCl4)-induced liver injury mice to assess specific toxicity in fibrotic liver tissue. Mice receiving LV-FAH alone (109 TU/mouse) or in combination with DEN/CCl4 presented clinically similar to control animals, with only slight reductions in total body weight gains over the study period (3.2- to 3.7-fold vs. 4.2-fold). There were no indications of toxicity attributed to administration of LV-FAH alone over the duration of this study. The known hepatotoxic combination of DEN/CCl4 induced fibrotic liver injury, and co-administration with LV-FAH was associated with exaggeration of some findings such as an increased liver:body weight ratio and progression to focal hepatocyte necrosis in some animals. Hepatocellular degeneration/regeneration was present in DEN/CCl4-dosed animals regardless of LV-FAH as evaluated by Ki-67 immunohistochemistry and circulating alpha fetoprotein levels, but there were no tumors identified in any tissue in any dose group. These data demonstrate the inherent safety of LV-FAH and support broader clinical development of lentiviral vectors for in vivo administration.
Collapse
Affiliation(s)
- Robert Allen Kaiser
- Midwest Fetal Care Center, Children's Hospital of Minnesota, Minneapolis, Minnesota
- Mayo Clinic, Department of Surgery Research, Rochester, Minnesota
| | | | - Kari Lynn Allen
- Mayo Clinic, Department of Surgery Research, Rochester, Minnesota
| | | | - Zeji Du
- Mayo Clinic, Department of Surgery Research, Rochester, Minnesota
| | | | - Joseph Benjamin Lillegard
- Midwest Fetal Care Center, Children's Hospital of Minnesota, Minneapolis, Minnesota
- Mayo Clinic, Department of Surgery Research, Rochester, Minnesota
- Pediatric Surgical Associates, Minneapolis, Minnesota
| |
Collapse
|
47
|
Norman BP, Davison AS, Ross GA, Milan AM, Hughes AT, Sutherland H, Jarvis JC, Roberts NB, Gallagher JA, Ranganath LR. A Comprehensive LC-QTOF-MS Metabolic Phenotyping Strategy: Application to Alkaptonuria. Clin Chem 2019; 65:530-539. [DOI: 10.1373/clinchem.2018.295345] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 01/28/2019] [Indexed: 11/06/2022]
Abstract
Abstract
BACKGROUND
Identification of unknown chemical entities is a major challenge in metabolomics. To address this challenge, we developed a comprehensive targeted profiling strategy, combining 3 complementary liquid chromatography quadrupole time-of-flight mass spectrometry (LC-QTOF-MS) techniques and in-house accurate mass retention time (AMRT) databases established from commercial standards. This strategy was used to evaluate the effect of nitisinone on the urinary metabolome of patients and mice with alkaptonuria (AKU). Because hypertyrosinemia is a known consequence of nitisinone therapy, we investigated the wider metabolic consequences beyond hypertyrosinemia.
METHODS
A total of 619 standards (molecular weight, 45–1354 Da) covering a range of primary metabolic pathways were analyzed using 3 liquid chromatography methods—2 reversed phase and 1 normal phase—coupled to QTOF-MS. Separate AMRT databases were generated for the 3 methods, comprising chemical name, formula, theoretical accurate mass, and measured retention time. Databases were used to identify chemical entities acquired from nontargeted analysis of AKU urine: match window theoretical accurate mass ±10 ppm and retention time ±0.3 min.
RESULTS
Application of the AMRT databases to data acquired from analysis of urine from 25 patients with AKU (pretreatment and after 3, 12, and 24 months on nitisinone) and 18 HGD−/− mice (pretreatment and after 1 week on nitisinone) revealed 31 previously unreported statistically significant changes in metabolite patterns and abundance, indicating alterations to tyrosine, tryptophan, and purine metabolism after nitisinone administration.
CONCLUSIONS
The comprehensive targeted profiling strategy described here has the potential of enabling discovery of novel pathways associated with pathogenesis and management of AKU.
Collapse
Affiliation(s)
- Brendan P Norman
- Musculoskeletal Biology I, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Andrew S Davison
- Musculoskeletal Biology I, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
- Department of Clinical Biochemistry and Metabolic Medicine, Liverpool Clinical Laboratories, Royal Liverpool University Hospitals Trust, Liverpool, UK
| | | | - Anna M Milan
- Musculoskeletal Biology I, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
- Department of Clinical Biochemistry and Metabolic Medicine, Liverpool Clinical Laboratories, Royal Liverpool University Hospitals Trust, Liverpool, UK
| | - Andrew T Hughes
- Musculoskeletal Biology I, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
- Department of Clinical Biochemistry and Metabolic Medicine, Liverpool Clinical Laboratories, Royal Liverpool University Hospitals Trust, Liverpool, UK
| | - Hazel Sutherland
- Musculoskeletal Biology I, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
- School of Exercise Science, Liverpool John Moores University, Liverpool, UK
| | - Jonathan C Jarvis
- School of Exercise Science, Liverpool John Moores University, Liverpool, UK
| | - Norman B Roberts
- Musculoskeletal Biology I, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - James A Gallagher
- Musculoskeletal Biology I, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Lakshminarayan R Ranganath
- Musculoskeletal Biology I, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
- Department of Clinical Biochemistry and Metabolic Medicine, Liverpool Clinical Laboratories, Royal Liverpool University Hospitals Trust, Liverpool, UK
| |
Collapse
|
48
|
Blundell J, Frisson S, Chakrapani A, Kearney S, Vijay S, MacDonald A, Gissen P, Hendriksz C, Olson A. Markers of cognitive function in individuals with metabolic disease: Morquio syndrome and tyrosinemia type III. Cogn Neuropsychol 2019; 35:120-147. [PMID: 29741470 DOI: 10.1080/02643294.2018.1443913] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
We characterized cognitive function in two metabolic diseases. MPS-IVa (mucopolysaccharidosis IVa, Morquio) and tyrosinemia type III individuals were assessed using tasks of attention, language and oculomotor function. MPS-IVa individuals were slower in visual search, but the display size effects were normal, and slowing was not due to long reaction times (ruling out slow item processing or distraction). Maintaining gaze in an oculomotor task was difficult. Results implicated sustained attention and task initiation or response processing. Shifting attention, accumulating evidence and selecting targets were unaffected. Visual search was also slowed in tyrosinemia type III, and patterns in visual search and fixation tasks pointed to sustained attention impairments, although there were differences from MPS-IVa. Language was impaired in tyrosinemia type III but not MPS-IVa. Metabolic diseases produced selective cognitive effects. Our results, incorporating new methods for developmental data and model selection, illustrate how cognitive data can contribute to understanding function in biochemical brain systems.
Collapse
Affiliation(s)
- James Blundell
- a School of Psychology , University of Birmingham , Birmingham , UK
| | - Steven Frisson
- a School of Psychology , University of Birmingham , Birmingham , UK
| | | | | | - Suresh Vijay
- b Birmingham Children's Hospital , Birmingham , UK
| | | | - Paul Gissen
- c Great Ormond Street Hospital , London , UK
| | - Chris Hendriksz
- d Steve Biko Academic Unit , University of Pretoria , Pretoria , South Africa
| | - Andrew Olson
- a School of Psychology , University of Birmingham , Birmingham , UK
| |
Collapse
|
49
|
Hickey RD, Nicolas CT, Allen K, Mao S, Elgilani F, Glorioso J, Amiot B, VanLith C, Guthman R, Du Z, Chen H, Harding CO, Kaiser RA, Nyberg SL, Lillegard JB. Autologous Gene and Cell Therapy Provides Safe and Long-Term Curative Therapy in A Large Pig Model of Hereditary Tyrosinemia Type 1. Cell Transplant 2018; 28:79-88. [PMID: 30477316 PMCID: PMC6322137 DOI: 10.1177/0963689718814188] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Orthotopic liver transplantation remains the only curative therapy for inborn errors of metabolism. Given the tremendous success for primary immunodeficiencies using ex-vivo gene therapy with lentiviral vectors, there is great interest in developing similar curative therapies for metabolic liver diseases. We have previously generated a pig model of hereditary tyrosinemia type 1 (HT1), an autosomal recessive disorder caused by deficiency of fumarylacetoacetate hydrolase (FAH). Using this model, we have demonstrated curative ex-vivo gene and cell therapy using a lentiviral vector to express FAH in autologous hepatocytes. To further evaluate the long-term clinical outcomes of this therapeutic approach, we continued to monitor one of these pigs over the course of three years. The animal continued to thrive off the protective drug NTBC, gaining weight appropriately, and maintaining sexual fecundity for the course of his life. The animal was euthanized 31 months after transplantation to perform a thorough biochemical and histological analysis. Biochemically, liver enzymes and alpha-fetoprotein levels remained normal and abhorrent metabolites specific to HT1 remained corrected. Liver histology showed no evidence of tumorigenicity and Masson's trichrome staining revealed minimal fibrosis and no evidence of cirrhosis. FAH-immunohistochemistry revealed complete repopulation of the liver by transplanted FAH-positive cells. A complete histopathological report on other organs, including kidney, revealed no abnormalities. This study is the first to demonstrate long-term safety and efficacy of hepatocyte-directed gene therapy in a large animal model. We conclude that hepatocyte-directed ex-vivo gene therapy is a rational choice for further exploration as an alternative therapeutic approach to whole organ transplantation for metabolic liver disease, including HT1.
Collapse
Affiliation(s)
- Raymond D Hickey
- 1 Department of Surgery, Mayo Clinic, Rochester, MN, USA.,2 Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Kari Allen
- 1 Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Shennen Mao
- 1 Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Jaime Glorioso
- 1 Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Bruce Amiot
- 1 Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Caitlin VanLith
- 1 Department of Surgery, Mayo Clinic, Rochester, MN, USA.,2 Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Rebekah Guthman
- 1 Department of Surgery, Mayo Clinic, Rochester, MN, USA.,2 Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Zeji Du
- 1 Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Harvey Chen
- 1 Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Cary O Harding
- 3 Department of Molecular and Medical Genetics, and Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Robert A Kaiser
- 1 Department of Surgery, Mayo Clinic, Rochester, MN, USA.,4 Midwest Fetal Care Center, Children's Hospital and Clinics of Minnesota, Minneapolis, MN, USA
| | - Scott L Nyberg
- 1 Department of Surgery, Mayo Clinic, Rochester, MN, USA.,5 William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, USA
| | - Joseph B Lillegard
- 1 Department of Surgery, Mayo Clinic, Rochester, MN, USA.,4 Midwest Fetal Care Center, Children's Hospital and Clinics of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
50
|
Davison AS, Norman BP, Smith EA, Devine J, Usher J, Hughes AT, Khedr M, Milan AM, Gallagher JA, Ranganath LR. Serum Amino Acid Profiling in Patients with Alkaptonuria Before and After Treatment with Nitisinone. JIMD Rep 2018; 41:109-117. [PMID: 29754208 DOI: 10.1007/8904_2018_109] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 03/28/2018] [Accepted: 04/13/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Alkaptonuria (AKU) is a rare inherited disorder of the tyrosine metabolic pathway. Our group is evaluating the use of the homogentisic acid-lowering agent nitisinone in patients with AKU. A major biochemical consequence of this treatment is hypertyrosinaemia. Herein we report the concentration of 20 serum amino acids over a 36-month period pre- and post-treatment with nitisinone. METHODS Fasting serum samples were collected at baseline (pre-nitisinone), 3 (2 mg nitisinone every other day), 6, 12, 24 and 36 (2 mg nitisinone daily) months. Amino acids were measured using the Biochrom 30 high-performance liquid chromatography cation exchange system with ninhydrin detection. RESULTS Fifty patients [21 female, mean age (±standard deviation) 54.1 (15.6) years (range 25-75); 29 male, mean age 49.3 (11.6) years (range 22-70 years)] were included. Following treatment mean tyrosine concentrations increased seven- to eight-fold (baseline, 69.8 μmol/L; 3 months, 670.7 μmol/L; 6 months, 666.4 μmol/L; 12 months, 692.9 μmol/L; 24 months, 649.4 μmol/L; 36 months, 724.8 μmol/L, p = <0.001 for all visits compared to baseline).At baseline mean phenylalanine, aspartic acid and arginine were outside the normal reference range. Following treatment the ratios of phenylalanine/tyrosine, phenylalanine/large neutral amino acids, arginine/branched chain amino acids and branched chain/aromatic amino acids decreased (p = <0.05), and the tyrosine/large neutral amino acid ratio increased (p = <0.0001). CONCLUSIONS Marked hypertyrosinaemia was observed following treatment with nitisinone. Noteworthy changes were also observed in the ratio of several amino acids following treatment with nitisinone suggesting that the availability of amino acids for neurotransmitter biosynthesis and liver function may be altered following treatment with nitisinone.
Collapse
Affiliation(s)
- A S Davison
- Liverpool Clinical Laboratories, Department of Clinical Biochemistry and Metabolic Medicine, Royal Liverpool University Hospitals Trust, Liverpool, UK.
- Bone and Joint Research Group, Musculoskeletal Biology, University of Liverpool, Liverpool, UK.
| | - B P Norman
- Bone and Joint Research Group, Musculoskeletal Biology, University of Liverpool, Liverpool, UK
| | - E A Smith
- Department of Clinical Biochemistry, Alder Hey Children's Hospital, Liverpool, UK
| | - J Devine
- Liverpool Clinical Laboratories, Department of Clinical Biochemistry and Metabolic Medicine, Royal Liverpool University Hospitals Trust, Liverpool, UK
| | - J Usher
- Liverpool Clinical Laboratories, Department of Clinical Biochemistry and Metabolic Medicine, Royal Liverpool University Hospitals Trust, Liverpool, UK
| | - A T Hughes
- Liverpool Clinical Laboratories, Department of Clinical Biochemistry and Metabolic Medicine, Royal Liverpool University Hospitals Trust, Liverpool, UK
- Bone and Joint Research Group, Musculoskeletal Biology, University of Liverpool, Liverpool, UK
| | - M Khedr
- Liverpool Clinical Laboratories, Department of Clinical Biochemistry and Metabolic Medicine, Royal Liverpool University Hospitals Trust, Liverpool, UK
- Bone and Joint Research Group, Musculoskeletal Biology, University of Liverpool, Liverpool, UK
| | - A M Milan
- Liverpool Clinical Laboratories, Department of Clinical Biochemistry and Metabolic Medicine, Royal Liverpool University Hospitals Trust, Liverpool, UK
- Bone and Joint Research Group, Musculoskeletal Biology, University of Liverpool, Liverpool, UK
| | - J A Gallagher
- Bone and Joint Research Group, Musculoskeletal Biology, University of Liverpool, Liverpool, UK
| | - L R Ranganath
- Liverpool Clinical Laboratories, Department of Clinical Biochemistry and Metabolic Medicine, Royal Liverpool University Hospitals Trust, Liverpool, UK
- Bone and Joint Research Group, Musculoskeletal Biology, University of Liverpool, Liverpool, UK
| |
Collapse
|