51
|
Leao Filho H, de Oliveira CV, Horvat N. Other types of diffuse liver disease: is there a way to do it? Abdom Radiol (NY) 2020; 45:3425-3443. [PMID: 32306241 DOI: 10.1007/s00261-020-02530-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
There are a variety of less common diffuse liver diseases that can be asymptomatic or cause severe liver dysfunction. For the majority of them, the association of clinical, laboratory, and imaging findings are needed to narrow the differential diagnosis. In this article, we will review and describe the rarer diffuse liver diseases including drug-related liver disease, inflammatory and infectious diseases, and deposition disorders such as amyloidosis, glycogen storage disease, Wilson's disease, and alpha-1 antitrypsin deficiency. Abdominal radiologists should be familiar with the imaging features of different types of diffuse liver diseases to help the multidisciplinary team involved in the treatment of these patients. The data related to some of these conditions are scarce and sometimes experimental, but we want to demonstrate to the reader the value of imaging techniques in their analysis and introduce the potential of new imaging methods.
Collapse
|
52
|
Çakar NE, Gezdirici A, Topuz HŞ, Önal H. Novel variants in Turkish patients with glycogen storage disease. Pediatr Int 2020; 62:1145-1150. [PMID: 32374048 DOI: 10.1111/ped.14286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 04/13/2020] [Accepted: 04/27/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Glycogen storage diseases (GSD) are disorders of autosomal recessive carbohydrate metabolism, characterized by glycogen accumulation. The liver and muscle tissue are commonly affected but patients may present with different clinical manifestations. The presence of glycogen can be demonstrated in biopsies and definitive diagnosis can be made by enzymatic or molecular analysis. The aim of this study was to determine specific gene mutations in our cases with GSD. METHODS Thirty-eight patients with clinical and laboratory diagnoses of GSD were studied. Thirty-two patients had undergone genetic analysis. In our study, a next-generation sequencing panel was used. RESULTS Five novel variants of uncertain significance (VUS), which were likely to be pathogenic, were detected in seven patients. Two new pathogenic variations of c.927delT (p.Phe309LeufsTer4) homozygous and c.44C>G (p.Ser15Ter) homozygous in the G6PC gene were detected in two GSD type Ia patients. In our two non-sibling GSD type III patients, c.1439T>G (p.Leu480Arg) homozygous novel-VUS was detected in the AGL gene. In our GSD type IV patient, c.1054G>C (p.Asp352His) homozygous novel-VUS was detected in the GBE1 gene. In GSD type VI, two sibling patients had a c.1454A>G (p.Asn485Ser) homozygous novel-VUS change in the PYGL gene. CONCLUSIONS We determined the gene mutations specific to cohorts in our cases with GSD. The novel pathogenic, likely pathogenic, and VUS changes identified will contribute to the relationship between the patients' clinical and laboratory findings.
Collapse
Affiliation(s)
- Nafiye Emel Çakar
- Division of Pediatric Metabolism, Okmeydanı Training and Research Hospital, University of Health Sciences, İstanbul, Turkey
| | - Alper Gezdirici
- Division of Medical Genetics, Kanuni Sultan Süleyman Training and Research Hospital, University of Health Sciences, İstanbul, Turkey
| | - Hanım Şeyma Topuz
- Division of Pediatric Metabolism, Kanuni Sultan Süleyman Training and Research Hospital, University of Health Sciences, İstanbul, Turkey
| | - Hasan Önal
- Division of Pediatric Metabolism, Kanuni Sultan Süleyman Training and Research Hospital, University of Health Sciences, İstanbul, Turkey
| |
Collapse
|
53
|
Nasrallah F, Hadj-Taieb S, Chehida AB, Jelassi A, Ben Massoued S, Charfi M, Zidi W, Amri F, Helel KB, Mejaoual H, Seboui H, Mahdhaoui N, Gargouri A, Monastiri K, Turki I, Cheour M, Sanhaji H, Tebib N, Feki M, Kaabachi N. Nonketotic Hyperglycinemia in Tunisia. Report upon a Series of 69 Patients. Neuropediatrics 2020; 51:349-353. [PMID: 32818969 DOI: 10.1055/s-0040-1712489] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
AIM The aim of the study is to report on epidemiological, clinical, and biochemical characteristics of nonketotic hyperglycinemia (NKH) in Tunisia. METHODS Patients diagnosed with NKH in Laboratory of Biochemistry at Rabta hospital (Tunis, Tunisia) between 1999 and 2018 were included. Plasma and cerebrospinal fluid (CSF) free amino acids were assessed by ion exchange chromatography. Diagnosis was based on family history, patient's clinical presentation and course, and increased CSF to plasma glycine ratio. RESULTS During 20 years, 69 patients were diagnosed with NKH, with 25 patients originating from Kairouan region. Estimated incidences were 1:55,641 in Tunisia and 1:9,684 in Kairouan. Consanguinity was found for 73.9% of the patients and 42% of the families have history of infantile death due to a disease of similar clinical course than the propositus. Clinical symptoms initiated within the first week of life in 75% of the patients and within the first 3 months in 95.7% ones. The phenotype was severe in 76.8% of the patients. Main symptoms were hypotonia, feeding difficulties, coma, apnea, and seizures. Most patients died within few days to months following diagnosis. CSF to plasma glycine ratio was increased in all patients. CSF and plasma glycine levels were negatively correlated with age of disease onset and severity. CONCLUSION NKH is quite frequent in Tunisia. Kairouan region has the highest NKH incidence rate, worldwide. However, due to lack of confirmatory enzymatic and genetic tests, NKH diagnosis was based on first-line biochemical tests. Characterization of causal mutations is needed for accurate diagnosis and prenatal diagnosis of this devastating life-threatening disease.
Collapse
Affiliation(s)
- Fahmi Nasrallah
- Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia.,Laboratory of Biochemistry, Rabta Hospital, Tunis, Tunisia.,Department of Pediatrics, Rabta Hospital, Tunis, Tunisia
| | - Sameh Hadj-Taieb
- Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia.,Laboratory of Biochemistry, Rabta Hospital, Tunis, Tunisia
| | - Amel Ben Chehida
- Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia.,Department of Pediatrics, Rabta Hospital, Tunis, Tunisia
| | - Awatef Jelassi
- Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia.,Laboratory of Biochemistry, Rabta Hospital, Tunis, Tunisia
| | - Sana Ben Massoued
- Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia.,Department of Pediatrics, Rabta Hospital, Tunis, Tunisia
| | - Manel Charfi
- Department of Neonatology, Hedi Chaker Hospital, Sfax, Tunisia
| | - Wiem Zidi
- Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia.,Laboratory of Biochemistry, Rabta Hospital, Tunis, Tunisia
| | - Fethi Amri
- Department of Pediatrics, Ibn Jazzar Hospital, Kairouan, Tunisia
| | - Khaled Ben Helel
- Department of Pediatrics, Ibn Jazzar Hospital, Kairouan, Tunisia
| | | | - Hassen Seboui
- Department of Neonatology, Farhat Hached Hospital, Sousse, Tunisia
| | - Nabiha Mahdhaoui
- Department of Neonatology, Farhat Hached Hospital, Sousse, Tunisia
| | | | - Kamel Monastiri
- Department of Neonatology, Fattouma Bourguiba Hospital, Monastir, Tunisia
| | - Ilhem Turki
- Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia.,Department of Child Neurology, National Institute of Neurology, Tunis, Tunisia
| | - Myriam Cheour
- Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia.,Department of Neonatology, Center of Maternity and Neonatology of Tunis, Tunis, Tunisia
| | - Haifa Sanhaji
- Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia.,Laboratory of Biochemistry, Rabta Hospital, Tunis, Tunisia
| | - Neji Tebib
- Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia.,Department of Pediatrics, Rabta Hospital, Tunis, Tunisia
| | - Moncef Feki
- Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia.,Laboratory of Biochemistry, Rabta Hospital, Tunis, Tunisia
| | - Naziha Kaabachi
- Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia.,Laboratory of Biochemistry, Rabta Hospital, Tunis, Tunisia
| |
Collapse
|
54
|
A Great Catch for Investigating Inborn Errors of Metabolism-Insights Obtained from Zebrafish. Biomolecules 2020; 10:biom10091352. [PMID: 32971894 PMCID: PMC7564250 DOI: 10.3390/biom10091352] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/18/2020] [Accepted: 09/19/2020] [Indexed: 12/14/2022] Open
Abstract
Inborn errors of metabolism cause abnormal synthesis, recycling, or breakdown of amino acids, neurotransmitters, and other various metabolites. This aberrant homeostasis commonly causes the accumulation of toxic compounds or depletion of vital metabolites, which has detrimental consequences for the patients. Efficient and rapid intervention is often key to survival. Therefore, it requires useful animal models to understand the pathomechanisms and identify promising therapeutic drug targets. Zebrafish are an effective tool to investigate developmental mechanisms and understanding the pathophysiology of disorders. In the past decades, zebrafish have proven their efficiency for studying genetic disorders owing to the high degree of conservation between human and zebrafish genes. Subsequently, several rare inherited metabolic disorders have been successfully investigated in zebrafish revealing underlying mechanisms and identifying novel therapeutic targets, including methylmalonic acidemia, Gaucher’s disease, maple urine disorder, hyperammonemia, TRAPPC11-CDGs, and others. This review summarizes the recent impact zebrafish have made in the field of inborn errors of metabolism.
Collapse
|
55
|
A Novel, Recurrent, 3.6-kb Deletion in the PYGL Gene Contributes to Glycogen Storage Disease Type VI. J Mol Diagn 2020; 22:1373-1382. [PMID: 32961316 DOI: 10.1016/j.jmoldx.2020.08.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 07/20/2020] [Accepted: 08/26/2020] [Indexed: 11/20/2022] Open
Abstract
The PYGL gene is the only established gene known to cause glycogen storage disease type VI (GSD6), which is a rare autosomal recessive disorder associated with hepatomegaly, elevated levels of hepatic transaminases, and hypoglycemia. Extended bioinformatics analysis was performed on the exome sequencing data of 5 patients who were clinically diagnosed as having or highly suspected of having GSD, and a single heterozygous pathogenic or likely pathogenic or rare variant of uncertain significance single-nucleotide variant was identified on the PYGL gene. A recurrent, novel, 3.6-kb deletion involving exons 14 to 17 of PYGL was identified in three of the five patients. Together with the two novel and one established stop-gain SNVs, they were diagnosed as compounds heterozygous of PYGL variants and confirmed as GSD6. The detected 3.6-kb deletion was further screened in a Chinese cohort of 31,317 individuals without hepatic abnormalities, and 10 carriers were identified, showing an allele frequency of 0.016%. Compared with the previously established 47 PYGL pathogenic or likely pathogenic SNVs, the novel pathogenic deletion had the second highest allele frequency among the population. This recurrent, novel, 3.6-kb deletion improved the molecular diagnostic rate of the GSD6. The relatively high frequency of the variant suggests that it is a potential mutation hotspot in patients with GSD6.
Collapse
|
56
|
Luciani M, Gritti A, Meneghini V. Human iPSC-Based Models for the Development of Therapeutics Targeting Neurodegenerative Lysosomal Storage Diseases. Front Mol Biosci 2020; 7:224. [PMID: 33062642 PMCID: PMC7530250 DOI: 10.3389/fmolb.2020.00224] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 08/10/2020] [Indexed: 01/30/2023] Open
Abstract
Lysosomal storage diseases (LSDs) are a group of rare genetic conditions. The absence or deficiency of lysosomal proteins leads to excessive storage of undigested materials and drives secondary pathological mechanisms including autophagy, calcium homeostasis, ER stress, and mitochondrial abnormalities. A large number of LSDs display mild to severe central nervous system (CNS) involvement. Animal disease models and post-mortem tissues partially recapitulate the disease or represent the final stage of CNS pathology, respectively. In the last decades, human models based on induced pluripotent stem cells (hiPSCs) have been extensively applied to investigate LSD pathology in several tissues and organs, including the CNS. Neural stem/progenitor cells (NSCs) derived from patient-specific hiPSCs (hiPS-NSCs) are a promising tool to define the effects of the pathological storage on neurodevelopment, survival and function of neurons and glial cells in neurodegenerative LSDs. Additionally, the development of novel 2D co-culture systems and 3D hiPSC-based models is fostering the investigation of neuron-glia functional and dysfunctional interactions, also contributing to define the role of neurodevelopment and neuroinflammation in the onset and progression of the disease, with important implications in terms of timing and efficacy of treatments. Here, we discuss the advantages and limits of the application of hiPS-NSC-based models in the study and treatment of CNS pathology in different LSDs. Additionally, we review the state-of-the-art and the prospective applications of NSC-based therapy, highlighting the potential exploitation of hiPS-NSCs for gene and cell therapy approaches in the treatment of neurodegenerative LSDs.
Collapse
Affiliation(s)
- Marco Luciani
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vasco Meneghini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
57
|
Murgasova L, Jurovcik M, Jesina P, Malinova V, Bloomfield M, Zeman J, Magner M. Otorhinolaryngological manifestations in 61 patients with mucopolysaccharidosis. Int J Pediatr Otorhinolaryngol 2020; 135:110137. [PMID: 32502916 DOI: 10.1016/j.ijporl.2020.110137] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 11/26/2022]
Abstract
OBJECTIVES The mucopolysaccharidoses (MPS) are inherited lysosomal storage disorders with multisystemic and highly variable clinical manifestation. ENT symptoms are common and early signs of MPS. The most common ENT diagnoses are chronic/recurrent rhinosinusitis, acute otitis media, otitis media with effusion, hearing loss and airway obstruction. METHODS A single-centre retrospective chart review of 61 patients (36 M/25F) with different MPS subtypes (MPS I (n = 15), MPS II (n = 10), MPS III (n = 17), MPS IV (n = 15) and MPS VI (n = 4)) was conducted. The age of ENT presentation and frequency of ENT symptoms, surgeries and their distribution among MPS subtypes was studied. The relationship between ENT presentation, first ENT surgery and the age of diagnosis was also evaluated. RESULTS Median age at the first ENT manifestation was 2.8 years, median age at MPS diagnosis 4.1 years. The great majority of patients (90%) manifested at least one ENT diagnosis; often before the diagnosis of MPS (75%). Chronic/recurrent rhinosinusitis was the most prevalent ENT diagnosis (77%), followed by upper airway obstruction (65%) and hearing loss (53%). Chronic/recurrent rhinosinusitis was the first ENT symptom to appear (median age 2.2 years), followed by otitis media with effusion (3.7 years) and hearing loss (4.5 years). At least one ENT surgery was performed in 57% of patients; in 69% before MPS diagnosis was established. Median age of the first ENT surgery was 4.1 years. ENT symptoms and surgical procedures were earliest present in MPS II. CONCLUSIONS Our study documents high and early occurrence of various otolaryngologic symptoms in MPS and thus highlights the role of ENT specialist in prompt diagnosis of these rare diseases and their long-term management.
Collapse
Affiliation(s)
- Lenka Murgasova
- Department of Pediatrics and Adolescent Medicine, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic; Department of ENT, Motol University Hospital and Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Michal Jurovcik
- Department of ENT, Motol University Hospital and Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Pavle Jesina
- Department of Pediatrics and Adolescent Medicine, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Vera Malinova
- Department of Pediatrics and Adolescent Medicine, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marketa Bloomfield
- Department of Immunology, Motol University Hospital and Second Faculty of Medicine, Charles University, Prague, Czech Republic; Department of Pediatrics, Thomayer's Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jiri Zeman
- Department of Pediatrics and Adolescent Medicine, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Martin Magner
- Department of Pediatrics and Adolescent Medicine, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic; Department of Pediatrics, Thomayer's Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
58
|
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: 1.8] [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
|
59
|
Sen K, Castillo Pinto C, Gropman AL. Expanding Role of Proton Magnetic Resonance Spectroscopy: Timely Diagnosis and Treatment Initiation in Partial Ornithine Transcarbamylase Deficiency. J Pediatr Genet 2020; 10:77-80. [PMID: 33552645 DOI: 10.1055/s-0040-1709670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 03/11/2020] [Indexed: 12/21/2022]
Abstract
We report the case of a 3-year-old male patient who presented with a 3-day history of altered mental status, emesis, and abdominal pain in the setting of a viral illness. A rapid screening revealed a high ammonia level and after reviewing his proton magnetic resonance spectroscopy (1H MRS) which showed the classic triad of high glutamate, low choline, and myoinositol, a diagnosis of ornithine transcarbamylase deficiency (OTCD) was made within 6 hours of presentation. Therapy with sodium phenylbutyrate and sodium benzoate was initiated and patient was discharged after 3 days with no neurologic disability. Biochemical and molecular testing eventually confirmed the diagnosis. 1H MRS is a practical and fast neuroimaging modality that can aid in diagnosis of OTCD and enables faster initiation of treatment in acute settings.
Collapse
Affiliation(s)
- Kuntal Sen
- Division of Neurogenetics and Developmental Pediatrics, Center for Neuroscience and Behavioral Medicine, Children's National Hospital, Washington, District of Columbia, United States
| | - Carlos Castillo Pinto
- Center for Neuroscience and Behavioral Medicine, Children's National Hospital, Washington, District of Columbia, United States
| | - Andrea L Gropman
- Division of Neurogenetics and Developmental Pediatrics, Center for Neuroscience and Behavioral Medicine, Children's National Hospital, Washington, District of Columbia, United States
| |
Collapse
|
60
|
Garbade SF, Zielonka M, Mechler K, Kölker S, Hoffmann GF, Staufner C, Mengel E, Ries M. FDA orphan drug designations for lysosomal storage disorders - a cross-sectional analysis. PLoS One 2020; 15:e0230898. [PMID: 32267884 PMCID: PMC7141691 DOI: 10.1371/journal.pone.0230898] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 03/11/2020] [Indexed: 12/30/2022] Open
Abstract
Purpose To provide a quantitative clinical-regulatory insight into the status of FDA orphan drug designations for compounds intended to treat lysosomal storage disorders (LSDs). Methods Assessment of the drug pipeline through analysis of the FDA database for orphan drug designations with descriptive and comparative statistics. Results Between 1983 and 2019, 124 orphan drug designations were granted by the FDA for compounds intended to treat 28 lysosomal storage diseases. Orphan drug designations focused on Gaucher disease (N = 16), Pompe disease (N = 16), Fabry disease (N = 10), MPS II (N = 10), MPS I (N = 9), and MPS IIIA (N = 9), and included enzyme replacement therapies, gene therapies, and small molecules, and others. Twenty-three orphan drugs were approved for the treatment of 11 LSDs. Gaucher disease (N = 6), cystinosis (N = 5), Pompe disease (N = 3), and Fabry disease (N = 2) had multiple approvals, CLN2, LAL-D, MPS I, II, IVA, VI, and VII one approval each. This is an increase of nine more approved drugs and four more treatable LSDs (CLN2, MPS VII, LAL-D, and MPS IVA) since 2013. Mean time between orphan drug designation and FDA approval was 89.7 SD 55.00 (range 8–203, N = 23) months. Conclusions The drug development pipeline for LSDs is growing and evolving, with increased focus on diverse small-molecule targets and gene therapy. CLN2 was the first and only LSD with an approved therapy directly targeted to the brain. Newly approved products included “me-too”–enzymes and innovative compounds such as the first pharmacological chaperone for the treatment of Fabry disease.
Collapse
Affiliation(s)
- Sven F. Garbade
- Division of Pediatric Neurology and Metabolic Medicine, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Matthias Zielonka
- Division of Pediatric Neurology and Metabolic Medicine, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Konstantin Mechler
- Department of Child and Adolescent Psychiatry and Psychotherapy & Department of Addictive Behavior and Addiction Medicine, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Stefan Kölker
- Division of Pediatric Neurology and Metabolic Medicine, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Georg F. Hoffmann
- Division of Pediatric Neurology and Metabolic Medicine, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Christian Staufner
- Division of Pediatric Neurology and Metabolic Medicine, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Eugen Mengel
- SphinCS GmbH, Science for LSD, Hochheim, Germany
| | - Markus Ries
- Division of Pediatric Neurology and Metabolic Medicine, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
- Center for Rare Diseases, University Hospital Heidelberg, Heidelberg, Germany
- Center for Virtual Patients, Medical Faculty, University of Heidelberg, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
61
|
Al-Sadeq DW, Nasrallah GK. The Spectrum of Mutations of Homocystinuria in the MENA Region. Genes (Basel) 2020; 11:330. [PMID: 32245022 PMCID: PMC7140887 DOI: 10.3390/genes11030330] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/09/2020] [Accepted: 03/13/2020] [Indexed: 12/18/2022] Open
Abstract
Homocystinuria is an inborn error of metabolism due to the deficiency in cystathionine beta-synthase (CBS) enzyme activity. It leads to the elevation of both homocysteine and methionine levels in the blood and urine. Consequently, this build-up could lead to several complications such as nearsightedness, dislocated eye lenses, a variety of psychiatric and behavioral disorders, as well as vascular system complications. The prevalence of homocystinuria is around 1/200,000 births worldwide. However, its prevalence in the Gulf region, notably Qatar, is exceptionally high and reached 1:1800. To date, more than 191 pathogenic CBS mutations have been documented. The majority of these mutations were identified in Caucasians of European ancestry, whereas only a few mutations from African-Americans or Asians were reported. Approximately 87% of all CBS mutations are missense and do not target the CBS catalytic site, but rather result in unstable misfolded proteins lacking the normal biological function, designating them for degradation. The early detection of homocystinuria along with low protein and methionine-restricted diet is the best treatment approach for all types of homocystinuria patients. Yet, less than 50% of affected individuals show a significant reduction in plasma homocysteine levels after treatment. Patients who fail to lower the elevated homocysteine levels, through high protein-restricted diet or by B6 and folic acid supplements, are at higher risk for cardiovascular diseases, neurodegenerative diseases, neural tube defects, and other severe clinical complications. This review aims to examine the mutations spectrum of the CBS gene, the disease management, as well as the current and potential treatment approaches with a greater emphasis on studies reported in the Middle East and North Africa (MENA) region.
Collapse
Affiliation(s)
- Duaa W. Al-Sadeq
- Biomedical Research Center, Qatar University, P.O. Box 2713 Doha, Qatar
- College of Medicine, Member of QU Health, Qatar University, P.O. Box 2713 Doha, Qatar
| | - Gheyath K. Nasrallah
- Biomedical Research Center, Qatar University, P.O. Box 2713 Doha, Qatar
- Department of Biomedical Science, College of Health Sciences, Qatar University, P.O. Box 2713 Doha, Qatar
| |
Collapse
|
62
|
Abstract
Metabolic disorders are disruptions in nutrient metabolism or basic cellular processes that can result in severe neonatal crisis. Basic laboratory findings may reveal hypoglycemia, acidosis, or hyperammonemia, but may also be normal even in infants with severe metabolic decompensation. Breast milk or milk-based formulas often contain the nutrient that precipitates the metabolic crisis and may need to be held during acute illness. Instead, infants with suspected metabolic disorders should be administered a high glucose infusion rate with isotonic fluids to reverse catabolism. Combined advanced biochemical and molecular testing is often needed to identify specific metabolic disorders and guide ongoing treatment.
Collapse
Affiliation(s)
- Katie B Williams
- Department of Pediatrics, University of Wisconsin Hospital and Clinics, 1500 Highland Avenue, Madison, WI 53705, USA
| | - Patrice K Held
- Department of Pediatrics, Wisconsin State Laboratory of Hygiene, University of Wisconsin School of Medicine and Public Health, 465 Henry Mall, Madison, WI 53706, USA
| | - Jessica Scott Schwoerer
- Department of Pediatrics, Division of Genetics and Metabolism, University of Wisconsin Hospital and Clinics, 1500 Highland Avenue, Madison, WI 53705, USA.
| |
Collapse
|
63
|
Long-Term Outcomes of Early Enzyme Replacement Therapy for Mucopolysaccharidosis IV: Clinical Case Studies of Two Siblings. Diagnostics (Basel) 2020; 10:diagnostics10020108. [PMID: 32079294 PMCID: PMC7168314 DOI: 10.3390/diagnostics10020108] [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: 11/20/2019] [Revised: 02/04/2020] [Accepted: 02/05/2020] [Indexed: 12/15/2022] Open
Abstract
Enzyme replacement therapy (ERT) is one of the available therapies for mucopolysaccharidosis (MPS). This study presents a follow-up of two siblings with MPS IVA (Morquio A disease) that received ERT. Both siblings received weekly intravenous infusions of elosulfase alfa for 4.5 years. One sibling (patient 1, P1; male) started therapy at 54 months of age, and the other sibling (patient 2, P2; female) started at 11 months of age. ERT was well-tolerated. In comparison to P1, P2’s growth curves deviated less from the norm. The orthopedic deformities of P1 were more severe than those of P2 and required several surgical corrections. P1’s sleep test at 48 months revealed obstructive sleep apnea, while by the age of 102 months, parameters were normal. P2 never had sleep apnea. Only P1 demonstrated ear, nose, and throat clinical illnesses. In comparison to P1, P2’s physical function was better maintained. In conclusion, ERT was safe in both patients during a 4.5-year follow-up. Although the typical characteristics of this disease were similar in both patients, P1 had a complex clinical course in comparison to P2, which influenced function and quality of life. Therefore, in order to make the most of ERT, it may be more beneficial when initiated at a relatively young age.
Collapse
|
64
|
Prevalence and clinical profile of glycogen storage diseases in children from Western India. Clin Exp Hepatol 2020; 6:9-12. [PMID: 32166118 PMCID: PMC7062124 DOI: 10.5114/ceh.2020.93050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/14/2019] [Indexed: 11/17/2022] Open
Abstract
Aim of the study To determine the prevalence and clinical profile of glycogen storage diseases (GSD) in children in western India. Material and methods This retrospective analysis was conducted over a period of 7 years from 2006 to 2012. All children diagnosed with GSD on liver biopsy were included in the study. Their clinical history, examination, biochemical profile and outcome were noted. Results Of 751 children, 18 (2.4%) were clinically diagnosed with GSD. Mean age at presentation was 2.3 ±1.3 years. Male : female ratio was 1 : 1. The main presenting features were abdominal distension in 15 (83.3%) patients, hepatomegaly in all 18 (100%), splenomegaly in 11 (61.1%) and jaundice in 2 (11.1%) patients. Four (22.2%) patients had delayed development. Four (22.2%) patients were siblings to an older affected child. Only 1 (5.6%) patient had portal hypertension and 2 (11.1%) had ascites. Only 1 (5.6%) patient had elevated bilirubin levels, 17 (94.4%) had elevated serum glutamic-oxaloacetic transaminase (SGOT) and all 18 (100%) patients had elevated serum glutamic-pyruvic transaminase (SGPT) levels. Nine (69.3%) patients of the 13 tested had acidosis, 1 (9.1%) had elevated uric acid, 2 (11.1%) had neutropenia, 8 (44.4%) experienced hypoglycemia, 4 (22.2%) patients had nephromegaly and only 1 patient showed evidence of cirrhosis in the liver biopsy. Fifteen (83.3%) patients were short. Three out of 6 patients tested had hypertriglyceridemia (50%). One (5.6%) patient died, 9 (50%) patients were lost to followup and the remaining 8 (44.4%) patients continued regular follow-up. Conclusions Metabolic acidosis, hypertriglyceridemia, short stature, and hypoglycemia are major problems in children with GSD. Most of the patients are referred late at the time of presentation.
Collapse
|
65
|
Federhen A, Pasqualim G, de Freitas TF, Gonzalez EA, Trapp F, Matte U, Giugliani R. Estimated birth prevalence of mucopolysaccharidoses in Brazil. Am J Med Genet A 2020; 182:469-483. [PMID: 31926052 DOI: 10.1002/ajmg.a.61456] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 11/03/2019] [Accepted: 11/10/2019] [Indexed: 11/05/2022]
Abstract
Several studies have been published on the frequency of the mucopolysaccharidoses (MPS) in different countries. The objective of the present study was to estimate the birth prevalence (BP) of MPS in Brazil. MPS diagnosis registered at MPS-Brazil Network and in Instituto Vidas Raras were reviewed. BP was estimated by (a) the number of registered patients born between 1994 and 2015 was divided by the number of live births (LBs), and (b) a sample of 1,000 healthy individuals was tested for the most frequent variant in IDUA gene in MPS I (p.Trp402Ter) to estimate the frequency of heterozygosity and homozygosity. (a) The BP based on total number of LBs was (cases per 100,000 LBs): MPS overall: 1.25; MPS I: 0.24; MPS II: 0.37; MPS III: 0.21; MPS IV: 0.14; MPS VI: 0.28; MPS VII: 0.02. (b) The overall frequency of p.Trp402Ter was 0.002. Considering the frequency of heterozygotes for the p.Trp402Ter IDUA variant in the RS state, the frequency of this variant among MPS I patients and the relative frequency of the different MPSs, we estimated the birth prevalence of MPS in total and of each MPS type, as follows: MPS overall: 4.62; MPS I: 0.95; MPS II: 1.32; MPS III: 0.56; MPS IV: 0.57; MPS VI: 1.02; MPS VII: 0.05. This study provided original data about BP and relative frequency of the MPS types, in Brazil, based on the frequency of the commonest IDUA pathogenic variant and in the records of two large patient databases.
Collapse
Affiliation(s)
- Andressa Federhen
- Postgraduate Program in Child and Adolescent Health, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Gabriela Pasqualim
- Postraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | | | - Esteban Alberto Gonzalez
- Postraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Franciele Trapp
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Postgraduate Program in Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Ursula Matte
- Postgraduate Program in Child and Adolescent Health, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Postraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Department of Genetics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Roberto Giugliani
- Postgraduate Program in Child and Adolescent Health, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Postraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Postgraduate Program in Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Department of Genetics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Instituto Nacional de Genética Médica Populacional/INAGEMP, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
66
|
Kija E, Wilmshurst JM. Approach to a Child with Epilepsy. CLINICAL CHILD NEUROLOGY 2020:795-808. [DOI: 10.1007/978-3-319-43153-6_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
67
|
Ying S, Zhihua Z, Yucan Z, Yu J, Qian L, Bixia Z, Weixia C, Zhifeng L. Molecular Diagnosis of Panel-Based Next-Generation Sequencing Approach and Clinical Symptoms in Patients With Glycogen Storage Disease: A Single Center Retrospective Study. Front Pediatr 2020; 8:600446. [PMID: 33344388 PMCID: PMC7744419 DOI: 10.3389/fped.2020.600446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 11/05/2020] [Indexed: 01/01/2023] Open
Abstract
Aim: The aim of this study was to investigate the clinical utility of panel-based next-generation sequencing (NGS) in the diagnostic approach of glycogen storage disease (GSD). Methods: We performed a retrospective review of the 32 cases with suspected GSDs between April 2013 and November 2019 through panel-based NGS, clinical and biochemical data and long-term complications. Results: Of the 32 clinical cases, we identified 41 different variants, including 24 missense (58.5%), one synonymous (2.4%), three nonsense (8%), one splice (2.4%), four frameshift (9.8%), one deletion (2.4%), four insertions (9.8%), two deletion-insertion (4.9%) and one duplication(2.4%), of which 13(31.7%) were previously unreported in the literature. In addition, patients with different types of GSDs showed important differences in biochemical parameters (i.e., CK, rGGT, TG, and UA). Conclusions: The panel-based NGS played an important diagnostic role in the suspicious GSDs patients, especially in the mild phenotype and ruled out detectable pathologic conditions. Besides, differences between our GSDs patients reflect biochemical heterogeneity.
Collapse
Affiliation(s)
- Shen Ying
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhang Zhihua
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zheng Yucan
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jin Yu
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Lin Qian
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zheng Bixia
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Cheng Weixia
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Liu Zhifeng
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
68
|
Safety Study of Sodium Pentosan Polysulfate for Adult Patients with Mucopolysaccharidosis Type II. Diagnostics (Basel) 2019; 9:diagnostics9040226. [PMID: 31861164 PMCID: PMC6963688 DOI: 10.3390/diagnostics9040226] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/07/2019] [Accepted: 12/09/2019] [Indexed: 12/14/2022] Open
Abstract
Current therapies for the mucopolysaccharidoses (MPS) do not effectively address skeletal and neurological manifestations. Pentosan polysulfate (PPS) is an alternative treatment strategy that has been shown to improve bone architecture, mobility, and neuroinflammation in MPS animals. The aims of this study were to a) primarily establish the safety of weekly PPS injections in attenuated MPS II, b) assess the efficacy of treatment on MPS pathology, and c) define appropriate clinical endpoints and biomarkers for future clinical trials. Subcutaneous injections were administered to three male Japanese patients for 12 weeks. Enzyme replacement therapy was continued in two of the patients while they received PPS and halted for two months in one patient before starting PPS. During treatment, one patient experienced an elevation of alanine transaminase, and another patient experienced convulsions; however, these incidences were non-cumulative and unrelated to PPS administration, respectively. Overall, the drug was well-tolerated in all patients, and no serious drug-related adverse events were noted. Generally, PPS treatment led to an increase in several parameters of shoulder range of motion and decrease of the inflammatory cytokines, MIF and TNF-α, which are potential clinical endpoints and biomarkers, respectively. Changes in urine and serum glycosaminoglycans were inconclusive. Overall, this study demonstrates the safety of using PPS in adults with MPS II and suggests the efficacy of PPS on MPS pathology with the identification of potential clinical endpoints and biomarkers.
Collapse
|
69
|
Torres DDA, Barth AL, Valente MPDM, de Mello PP, Horovitz DDG. Otolaryngologists and the Early Diagnosis of Mucopolysaccharidoses: A Cross-Sectional Study. Diagnostics (Basel) 2019; 9:diagnostics9040187. [PMID: 31766106 PMCID: PMC6963463 DOI: 10.3390/diagnostics9040187] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/29/2019] [Accepted: 10/01/2019] [Indexed: 12/20/2022] Open
Abstract
Mucopolysaccharidoses (MPS) are a group of inborn errors of metabolism with an aggressive and usually fatal course. Therefore, early treatment is essential because the involvement of head and neck structures is almost always present in MPS. Our study aimed to retrospectively assess—via a chart review and a survey of caregivers—the history of ear, nose and throat (ENT) symptoms, the number of otolaryngology visits prior to diagnosis, and whether otolaryngologists diagnosed the disease in a cohort of MPS patients followed at an academic medical center. Twenty-three patients were evaluated. Age at diagnosis ranged from 0.2 to 33.0 years (median, 3.2 years). Prior to being diagnosed with MPS, 20/23 (87%) patients presented with at least one episode of otalgia, airway disorder, sleep disturbance, speech delay or suspected hearing loss. One patient had an adenotonsillectomy with paracentesis of tympanic membranes. Ten of the 23 patients (43%) were seen by an otolaryngologist before the diagnosis of MPS, none of which had the disease suspected during these visits. Notwithstanding limitations, our results suggest that increased awareness of MPS among otolaryngologists may allow for earlier diagnosis and better management of these patients.
Collapse
|
70
|
Baruteau J, Diez-Fernandez C, Lerner S, Ranucci G, Gissen P, Dionisi-Vici C, Nagamani S, Erez A, Häberle J. Argininosuccinic aciduria: Recent pathophysiological insights and therapeutic prospects. J Inherit Metab Dis 2019; 42:1147-1161. [PMID: 30723942 DOI: 10.1002/jimd.12047] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 12/20/2018] [Indexed: 12/30/2022]
Abstract
The first patients affected by argininosuccinic aciduria (ASA) were reported 60 years ago. The clinical presentation was initially described as similar to other urea cycle defects, but increasing evidence has shown overtime an atypical systemic phenotype with a paradoxical observation, that is, a higher rate of neurological complications contrasting with a lower rate of hyperammonaemic episodes. The disappointing long-term clinical outcomes of many of the patients have challenged the current standard of care and therapeutic strategy, which aims to normalize plasma ammonia and arginine levels. Interrogations have raised about the benefit of newborn screening or liver transplantation on the neurological phenotype. Over the last decade, novel discoveries enabled by the generation of new transgenic argininosuccinate lyase (ASL)-deficient mouse models have been achieved, such as, a better understanding of ASL and its close interaction with nitric oxide metabolism, ASL physiological role outside the liver, and the pathophysiological role of oxidative/nitrosative stress or excessive arginine treatment. Here, we present a collaborative review, which highlights these recent discoveries and novel emerging concepts about ASL role in human physiology, ASA clinical phenotype and geographic prevalence, limits of current standard of care and newborn screening, pathophysiology of the disease, and emerging novel therapies. We propose recommendations for monitoring of ASA patients. Ongoing research aims to better understand the underlying pathogenic mechanisms of the systemic disease to design novel therapies.
Collapse
Affiliation(s)
- Julien Baruteau
- UCL Great Ormond Street Institute of Child Health, NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
- Metabolic Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Carmen Diez-Fernandez
- Division of Metabolism and Children Research Centre (CRC), University Children's Hospital, Zurich, Switzerland
| | - Shaul Lerner
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israël
| | - Giusy Ranucci
- Division of Metabolism, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Paul Gissen
- UCL Great Ormond Street Institute of Child Health, NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
- Metabolic Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Carlo Dionisi-Vici
- Division of Metabolism, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Sandesh Nagamani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israël
| | - Johannes Häberle
- Division of Metabolism and Children Research Centre (CRC), University Children's Hospital, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP) and Neuroscience Center Zurich (ZNZ), Zurich, Switzerland
| |
Collapse
|
71
|
Abstract
Metabolomics uses advanced analytical chemistry techniques to enable the high-throughput characterization of metabolites from cells, organs, tissues, or biofluids. The rapid growth in metabolomics is leading to a renewed interest in metabolism and the role that small molecule metabolites play in many biological processes. As a result, traditional views of metabolites as being simply the "bricks and mortar" of cells or just the fuel for cellular energetics are being upended. Indeed, metabolites appear to have much more varied and far more important roles as signaling molecules, immune modulators, endogenous toxins, and environmental sensors. This review explores how metabolomics is yielding important new insights into a number of important biological and physiological processes. In particular, a major focus is on illustrating how metabolomics and discoveries made through metabolomics are improving our understanding of both normal physiology and the pathophysiology of many diseases. These discoveries are yielding new insights into how metabolites influence organ function, immune function, nutrient sensing, and gut physiology. Collectively, this work is leading to a much more unified and system-wide perspective of biology wherein metabolites, proteins, and genes are understood to interact synergistically to modify the actions and functions of organelles, organs, and organisms.
Collapse
Affiliation(s)
- David S Wishart
- Departments of Biological Sciences and Computing Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
72
|
Bower A, Imbard A, Benoist JF, Pichard S, Rigal O, Baud O, Schiff M. Diagnostic contribution of metabolic workup for neonatal inherited metabolic disorders in the absence of expanded newborn screening. Sci Rep 2019; 9:14098. [PMID: 31575911 PMCID: PMC6773867 DOI: 10.1038/s41598-019-50518-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 09/05/2019] [Indexed: 12/11/2022] Open
Abstract
Inherited metabolic disorders (IMDs) in neonates are a diagnostic and therapeutic challenge for the neonatologist, with the priority being to rapidly flag the treatable diseases. The objective of this study was to evaluate the contribution of targeted metabolic testing for diagnosing suspected IMDs on the basis of suggestive clinical setting or family history in neonates. We conducted an observational study over five years, from January 1st, 2010 to December 31, 2014 in the neonatal intensive care unit (NICU) at Robert Debré University Hospital, Paris, France. We assessed the number of neonates for whom a metabolic testing was performed, the indication for each metabolic test and the diagnostic yield of this selected metabolic workup for diagnosing an IMD. Metabolic testing comprised at least one of the following testings: plasma, urine or cerebrospinal fluid amino acids, urine organic acids, plasma acylcarnitine profile, and urine mucopolysaccharides and oligosaccharides. 11,301 neonates were admitted at the neonatal ICU during the study period. One hundred and ninety six neonates underwent metabolic testing. Eleven cases of IMDs were diagnosed. This diagnostic approach allowed the diagnosis, treatment and survival of 4 neonates (maple syrup urine disease, propionic acidemia, carnitine-acylcarnitine translocase deficiency and type 1 tyrosinemia). In total, metabolic testing was performed for 1.7% of the total number of neonates admitted in the NICU over the study period. These included 23% finally unaffected neonates with transient abnormalities, 5.6% neonates suffering from an identified IMD, 45.4% neonates suffering from a non-metabolic identified disease and 26% neonates with chronic abnormalities but for whom no final causal diagnosis could be made. In conclusion, as expected, such a metabolic targeted workup allowed the diagnosis of classical neonatal onset IMDs in symptomatic newborns. However, this workup remained normal or unspecific for 94.4% of the tested patients. It allowed excluding an IMD in 68.4% of the tested neonates. In spite of the high rate of normal results, such a strategy seems acceptable due to the severity of the symptoms and the need for immediate treatment when available in neonatal IMDs. However, its cost-effectiveness remains low especially in a clinically targeted population in a country where newborn screening is still unavailable for IMDs except for phenylketonuria in 2019.
Collapse
Affiliation(s)
- Alexandra Bower
- Neonatal intensive care department, Robert Debré University Hospital, APHP, Paris, 75019, France
- Reference Center for Inborn Errors of Metabolism, Robert Debré University Hospital, APHP, Paris, 75019, France
| | - Apolline Imbard
- Biochemistry Laboratory, Robert Debré University Hospital, APHP, Paris, France
- Paris Sud University, Chatenay Malabry, France
| | - Jean-François Benoist
- Biochemistry Laboratory, Robert Debré University Hospital, APHP, Paris, France
- Paris Sud University, Chatenay Malabry, France
| | - Samia Pichard
- Reference Center for Inborn Errors of Metabolism, Robert Debré University Hospital, APHP, Paris, 75019, France
| | - Odile Rigal
- Biochemistry Laboratory, Robert Debré University Hospital, APHP, Paris, France
| | - Olivier Baud
- Neonatal intensive care department, Robert Debré University Hospital, APHP, Paris, 75019, France
- UMR1141, PROTECT, INSERM, Université de Paris, Paris, 75019, France
| | - Manuel Schiff
- Reference Center for Inborn Errors of Metabolism, Robert Debré University Hospital, APHP, Paris, 75019, France.
- UMR1141, PROTECT, INSERM, Université de Paris, Paris, 75019, France.
| |
Collapse
|
73
|
Glinton KE, Elsea SH. Untargeted Metabolomics for Autism Spectrum Disorders: Current Status and Future Directions. Front Psychiatry 2019; 10:647. [PMID: 31551836 PMCID: PMC6746843 DOI: 10.3389/fpsyt.2019.00647] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 08/12/2019] [Indexed: 12/20/2022] Open
Abstract
Autism spectrum disorders (ASDs) are a group of neurodevelopment disorders characterized by childhood onset deficits in social communication and interaction. Although the exact etiology of most cases of ASDs is unknown, a portion has been proposed to be associated with various metabolic abnormalities including mitochondrial dysfunction, disorders of cholesterol metabolism, and folate abnormalities. Targeted biochemical testing like plasma amino acid and acylcarnitine profiles have demonstrated limited utility in helping to diagnose and manage such patients. Untargeted metabolomics has emerged, however, as a promising tool in screening for underlying biochemical abnormalities and managing treatment and as a means of investigating possible novel biomarkers for the disorder. Here, we review the principles and methodology behind untargeted metabolomics, recent pilot studies utilizing this technology, and areas in which it may be integrated into the care of children with this disorder in the future.
Collapse
Affiliation(s)
- Kevin E. Glinton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Sarah H. Elsea
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
74
|
Bijarnia-Mahay S, Kapoor S. Testing Modalities for Inborn Errors of Metabolism — What a Clinician Needs to Know? Indian Pediatr 2019. [DOI: 10.1007/s13312-019-1637-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
75
|
Poothrikovil RP, Al Thihli K, Al Futaisi A, Al Murshidi F. Nonketotic Hyperglycinemia: Two Case Reports and Review. Neurodiagn J 2019; 59:142-151. [PMID: 31433733 DOI: 10.1080/21646821.2019.1645549] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Nonketotic hyperglycinemia (NKH) or glycine encephalopathy is an autosomal recessive disorder of glycine metabolism resulting in an excessive accumulation of glycine in all body tissues, including the central nervous system. It is caused by a biochemical defect in the glycine cleavage system and considered as a rare disorder with an estimated prevalence of 1:60,000. The neonatal form presents in the first few days of life with progressive encephalopathy, hypotonia, myoclonic jerks, hiccups, seizures, rapid progression to coma and often death due to central apnea. Surviving infants often have severe developmental delay and refractory seizures. Atypical forms of NKH present with heterogeneous and nonspecific disease course. Classical glycine encephalopathy usually carries a very poor prognosis. We describe two neonates who presented with neonatal encephalopathy, apnea, and progressive lethargy. Increased CSF glycine level along with an elevated CSF to plasma glycine ratio was suggestive of classic NKH. Burst suppression EEG and agenesis of the corpus callosum were supportive findings. Evolution of the EEG patterns and course of the disease are discussed in detail. Transient phases of clinical stabilization and normalized plasma biochemical results may not necessarily reflect the actual encephalopathic process. Serial EEGs are helpful to assess the efficacy of treatment and to modify the therapeutic approach.
Collapse
Affiliation(s)
- Rajesh P Poothrikovil
- Department of Clinical PhysiologySultan Qaboos University Hospital , Muscat , Sultanate of Oman
| | - Khalid Al Thihli
- Department of Clinical GeneticsSultan Qaboos University Hospital , Muscat , Sultanate of Oman
| | - Amna Al Futaisi
- Department of Child HealthSultan Qaboos University Hospital , Muscat , Sultanate of Oman
| | - Fathiya Al Murshidi
- Department of Clinical GeneticsSultan Qaboos University Hospital , Muscat , Sultanate of Oman
| |
Collapse
|
76
|
Tanacan A, Gurbuz BB, Aydin E, Erden M, Coskun T, Beksac MS. Prenatal Diagnosis of Organic Acidemias at a Tertiary Center. Balkan J Med Genet 2019; 22:29-34. [PMID: 31523617 PMCID: PMC6714333 DOI: 10.2478/bjmg-2019-0003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The aim of this study was to share our experience in the prenatal diagnosis (PND) of organic acidemias (OAs) in our clinic. This study consisted of 10 cases in whom an invasive prenatal diagnostic test (IPNDT) was performed by a single physician for the PND of OAs. Median maternal age, parity, gestational week of IPNDT, prenatal test indications, OA types, method of IPNDT, IPNDT results and gestational outcomes were evaluated. Targeted mutation analysis was performed in fetal DNA for the specific mutations by using polymerase chain reaction (PCR) and direct Sanger sequencing. The diagnosis was confirmed by genetic targeted mutation analysis after birth. Median maternal age, parity and gestational week of IPNDT values were 30 (range 21-35), one (range 0-4) and 11.5 (range 11-17), respectively. Indications for IPNDT were mother being a carrier of the disease for one case (10.0%) and at least one child with OA in the family for nine cases (90.0%). Organic acidemia types investigated were maple syrup urine disease (MSUD), methylmalonic acidemia (MMA) and isovaleric acidemia (IVA) in five (50.0%), three (30.0%) and two (20.0%) patients, respectively. Chorion villus sampling (CVS) was done in seven (70.0%) patients and amniocentesis was performed in three (30.0%) patients. Eight fetuses (80.0%) were found to be healthy and two fetuses (20.0%) were found to be affected (one case with IVA and one case with MMA). The two pregnancies (20.0%) with affected fetuses were terminated. Prenatal diagnosis of OAs is critical. Appropriate prenatal counseling should be given to families with known risk factors.
Collapse
Affiliation(s)
- A Tanacan
- Department of Obstetrics and Gynecology, Division of Perinatology, Hacettepe University Hospital, Ankara, Turkey
| | - BB Gurbuz
- Department of Pediatrics, Division of Pediatric Metabolism, Hacettepe University Hospital, Ankara, Turkey
| | - E Aydin
- Department of Obstetrics and Gynecology, Division of Perinatology, Hacettepe University Hospital, Ankara, Turkey
| | - M Erden
- Department of Obstetrics and Gynecology, Division of Perinatology, Hacettepe University Hospital, Ankara, Turkey
| | - T Coskun
- Department of Pediatrics, Division of Pediatric Metabolism, Hacettepe University Hospital, Ankara, Turkey
| | - MS Beksac
- Department of Obstetrics and Gynecology, Division of Perinatology, Hacettepe University Hospital, Ankara, Turkey
| |
Collapse
|
77
|
Almási T, Guey LT, Lukacs C, Csetneki K, Vokó Z, Zelei T. Systematic literature review and meta-analysis on the epidemiology of methylmalonic acidemia (MMA) with a focus on MMA caused by methylmalonyl-CoA mutase (mut) deficiency. Orphanet J Rare Dis 2019; 14:84. [PMID: 31023387 PMCID: PMC6485056 DOI: 10.1186/s13023-019-1063-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/08/2019] [Indexed: 12/13/2022] Open
Abstract
Methylmalonic acidemia/aciduria (MMA) is a genetically heterogeneous group of inherited metabolic disorders biochemically characterized by the accumulation of methylmalonic acid. Isolated MMA is primarily caused by the deficiency of methylmalonyl-CoA mutase (MMA mut; EC 5.4.99.2). A systematic literature review and a meta-analysis were undertaken to assess and compile published epidemiological data on MMA with a focus on the MMA mut subtype (OMIM #251000). Of the 1114 identified records, 227 papers were assessed for eligibility in full text, 48 articles reported on disease epidemiology, and 39 articles were included into the quantitative synthesis. Implementation of newborn screening in various countries has allowed for the estimation of birth prevalence of MMA and its isolated form. Meta-analysis pooled point estimates of MMA (all types) detection rates were 0.79, 1.12, 1.22 and 6.04 per 100,000 newborns in Asia-Pacific, Europe, North America and the Middle East and North Africa (MENA) regions, respectively. The detection rate of isolated MMA was < 1 per 100,000 newborns in all regions with the exception of MENA where it approached 6 per 100,000 newborns. Few studies published data on the epidemiology of MMA mut, therefore no meta-analysis could have been performed on this subtype. Most of the identified papers reported birth prevalence estimates below 1 per 100,000 newborns for MMA mut. The systematic literature review clearly demonstrates that MMA and its subtypes are ultra-rare disorders.
Collapse
Affiliation(s)
- Tímea Almási
- Syreon Research Institute, Mexikói str. 65/A, Budapest, H-1142, Hungary.
| | | | | | - Kata Csetneki
- Syreon Research Institute, Mexikói str. 65/A, Budapest, H-1142, Hungary
| | - Zoltán Vokó
- Syreon Research Institute, Mexikói str. 65/A, Budapest, H-1142, Hungary.,Department of Health Policy & Health Economics, Eötvös Loránd University, Budapest, Hungary
| | - Tamás Zelei
- Syreon Research Institute, Mexikói str. 65/A, Budapest, H-1142, Hungary
| |
Collapse
|
78
|
Doherty C, Stapleton M, Piechnik M, Mason RW, Mackenzie WG, Yamaguchi S, Kobayashi H, Suzuki Y, Tomatsu S. Effect of enzyme replacement therapy on the growth of patients with Morquio A. J Hum Genet 2019; 64:625-635. [PMID: 31019230 DOI: 10.1038/s10038-019-0604-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/22/2019] [Accepted: 04/08/2019] [Indexed: 01/07/2023]
Abstract
Mucopolysaccharidosis IVA (MPS IVA) is a degenerative systemic skeletal dysplasia, in which children exhibit marked short stature and become physically handicapped. This study evaluated the growth patterns of patients treated with enzyme replacement therapy (ERT), compared with those of untreated patients. Cross-sectional and longitudinal data of heights and weights were collected from 128 MPS IVA patients and compared with the growth charts of MPS IVA. Twelve patients (six males, six females) starting ERT before 5 years old were treated for at least 2 years. Six out of 12 patients (50%) with ERT over 2 years stopped growing between 94 and 98 cm (mean height of 95.1 ± 2.2 cm) from 5.0 years to 9.0 years of age (mean age of 6.2 ± 1.6 years). The other patients, except one attenuated case, exhibited a marked slow growth velocity from 3.6 years to 7.7 years. Treated and untreated patients with severe phenotype reached their final heights by ~10 years of age. Patients treated with ERT exhibited a reduced pubertal growth spurt analogous to their untreated counterparts, which contributes to the marked short stature associated with MPS IVA. Compared with the growth charts for untreated patients, patients treated with ERT did not show any significant increase in growth in any age group. Overall, ERT-treated patients do not experience growth improvement and continue to exhibit poor growth despite early ERT intervention before 5 years of age. These findings indicate that current intravenous ERT is ineffective at correcting abnormal growth in MPS IVA.
Collapse
Affiliation(s)
- Caitlin Doherty
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA.,University of Delaware, Newark, DE, USA
| | - Molly Stapleton
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA.,University of Delaware, Newark, DE, USA
| | - Matthew Piechnik
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA.,University of Delaware, Newark, DE, USA
| | - Robert W Mason
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA.,University of Delaware, Newark, DE, USA
| | | | - Seiji Yamaguchi
- Department of Pediatrics, Shimane University, Shimane, Japan
| | | | - Yasuyuki Suzuki
- Medical Education Development Center, Gifu University, Gifu, Japan
| | - Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA. .,Department of Pediatrics, Shimane University, Shimane, Japan. .,Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
79
|
P4 medicine for epilepsy and intellectual disability: nutritional therapy for inherited metabolic disease. Emerg Top Life Sci 2019; 3:75-95. [PMID: 33523196 DOI: 10.1042/etls20180180] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/20/2019] [Accepted: 02/25/2019] [Indexed: 12/16/2022]
Abstract
Early identification and treatment of inherited metabolic diseases (IMDs) are essential to prevent and minimize intellectual disability (ID) and epilepsy. The oldest form of treatment, nutritional modulation, has proved beneficial for many IMDs. These conditions represent a promising model for P4 medicine - predictive, preventive, personalized, and participatory - specifically through the interpretation of individual genetic, pathophysiological, and clinical characteristics. More than 1000 IMDs have been described, and for these different nutritional modulation strategies are applied, varying from substrate reduction, supplementation of vitamins for catalyzation of enzymatic reactions or supplementation of amino acids or other nutrients, to substitution for deficient or inactivated products. This review provides an overview of all IMDs presenting with epilepsy and/or ID amenable to nutritional modulation; these are 85 in number, belonging to 27 categories. Therapeutic strategies include protein-restricted diet, ketogenic diet, fat-restricted diet, lactose-restricted diet; supplementation of amino acids, carbohydrates, or others; and supplementation of vitamins or cofactors as well as a sick-day protocol. Nutritional therapies are generally safe, affordable, and accessible, but compliance is an issue. Three different types of response exist: (1) a positive effect on seizure control and/or psychomotor development; (2) efficacy in prevention of decompensation but ongoing damage occurs; and (3) insufficient insights or evidence to establish the treatment as effective. For the latter category, we describe pyridoxine-dependent epilepsy as a case vignette for P4 medicine, discuss the benefits and challenges of nutritional modulation in IMDs, and outline novel approaches and solutions.
Collapse
|
80
|
|
81
|
Almási T, Guey LT, Lukacs C, Csetneki K, Vokó Z, Zelei T. Systematic literature review and meta-analysis on the epidemiology of propionic acidemia. Orphanet J Rare Dis 2019; 14:40. [PMID: 30760309 PMCID: PMC6375193 DOI: 10.1186/s13023-018-0987-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 12/21/2018] [Indexed: 02/06/2023] Open
Abstract
Propionic acidemia (PA, OMIM #606054) is a serious, life-threatening, inherited, metabolic disorder caused by the deficiency of the mitochondrial enzyme propionyl-coenzyme A (CoA) carboxylase (EC 6.4.1.3). The primary objective of this study was to conduct a systematic literature review and meta-analysis on the epidemiology of PA. The literature search was performed covering Medline, Embase, Cochrane Database of Systematic Reviews, CRD Database, Academic Search Complete, CINAHL and PROSPERO databases. Websites of rare disease organizations were also searched for eligible studies. Of the 2338 identified records, 188 articles were assessed for eligibility in full text, 43 articles reported on disease epidemiology, and 31 studies were included into the quantitative synthesis. Due to the rarity of PA, broadly targeted population-based prevalence studies are not available. Nonetheless, implementation of newborn screening programs has allowed the estimation of the birth prevalence data of PA across multiple geographic regions. The pooled point estimates indicated detection rates of 0.29; 0.33; 0.33 and 4.24 in the Asia-Pacific, Europe, North America and the Middle East and North Africa (MENA) regions, respectively. Our systematic literature review and meta-analysis confirm that PA is an ultra-rare disorder, with similar detection rates across all regions with the exception of the MENA region where the disease, similar to other inherited metabolic disorders, is more frequent.
Collapse
Affiliation(s)
- Tímea Almási
- Syreon Research Institute, Mexikói str. 65/A, Budapest, H-1142, Hungary.
| | | | | | - Kata Csetneki
- Syreon Research Institute, Mexikói str. 65/A, Budapest, H-1142, Hungary
| | - Zoltán Vokó
- Syreon Research Institute, Mexikói str. 65/A, Budapest, H-1142, Hungary.,Department of Health Policy & Health Economics, Eötvös Loránd University, Budapest, Hungary
| | - Tamás Zelei
- Syreon Research Institute, Mexikói str. 65/A, Budapest, H-1142, Hungary
| |
Collapse
|
82
|
Substrate reduction therapy for inborn errors of metabolism. Emerg Top Life Sci 2019; 3:63-73. [PMID: 33523197 PMCID: PMC7289018 DOI: 10.1042/etls20180058] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 01/02/2019] [Accepted: 01/09/2019] [Indexed: 12/13/2022]
Abstract
Inborn errors of metabolism (IEM) represent a growing group of monogenic disorders each associated with inherited defects in a metabolic enzyme or regulatory protein, leading to biochemical abnormalities arising from a metabolic block. Despite the well-established genetic linkage, pathophysiology and clinical manifestations for many IEMs, there remains a lack of transformative therapy. The available treatment and management options for a few IEMs are often ineffective or expensive, incurring a significant burden to individual, family, and society. The lack of IEM therapies, in large part, relates to the conceptual challenge that IEMs are loss-of-function defects arising from the defective enzyme, rendering pharmacologic rescue difficult. An emerging approach that holds promise and is the subject of a flurry of pre-/clinical applications, is substrate reduction therapy (SRT). SRT addresses a common IEM phenotype associated with toxic accumulation of substrate from the defective enzyme, by inhibiting the formation of the substrate instead of directly repairing the defective enzyme. This minireview will summarize recent highlights towards the development of emerging SRT, with focussed attention towards repurposing of currently approved drugs, approaches to validate novel targets and screen for hit molecules, as well as emerging advances in gene silencing as a therapeutic modality.
Collapse
|
83
|
Sobrido MJ, Bauer P, de Koning T, Klopstock T, Nadjar Y, Patterson MC, Synofzik M, Hendriksz CJ. Recommendations for patient screening in ultra-rare inherited metabolic diseases: what have we learned from Niemann-Pick disease type C? Orphanet J Rare Dis 2019; 14:20. [PMID: 30665446 PMCID: PMC6341610 DOI: 10.1186/s13023-018-0985-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 12/21/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Rare and ultra-rare diseases (URDs) are often chronic and life-threatening conditions that have a profound impact on sufferers and their families, but many are notoriously difficult to detect. Niemann-Pick disease type C (NP-C) serves to illustrate the challenges, benefits and pitfalls associated with screening for ultra-rare inborn errors of metabolism (IEMs). A comprehensive, non-systematic review of published information from NP-C screening studies was conducted, focusing on diagnostic methods and study designs that have been employed to date. As a key part of this analysis, data from both successful studies (where cases were positively identified) and unsuccessful studies (where the chosen approach failed to identify any cases) were included alongside information from our own experiences gained from the planning and execution of screening for NP-C. On this basis, best-practice recommendations for ultra-rare IEM screening are provided. Twenty-six published screening studies were identified and categorised according to study design into four groups: 1) prospective patient cohort and family-based secondary screenings (18 studies); 2) analyses of archived 'biobank' materials (one study); 3) medical chart review and bioinformatics data mining (five studies); and 4) newborn screening (two studies). NPC1/NPC2 sequencing was the most common primary screening method (Sanger sequencing in eight studies and next-generation sequencing [gene panel or exome sequencing] in five studies), followed by biomarker analyses (usually oxysterols) and clinical surveillance. CONCLUSIONS Historically, screening for NP-C has been based on single-patient studies, small case series, and targeted cohorts, but the emergence of new diagnostic methods over the last 5-10 years has provided opportunities to screen for NP-C on a larger scale. Combining clinical, biomarker and genetic diagnostic methods represents the most effective way to identify NP-C cases, while reducing the likelihood of misdiagnosis. Our recommendations are intended as a guide for planning screening protocols for ultra-rare IEMs in general.
Collapse
Affiliation(s)
- María-Jesús Sobrido
- Neurogenetics Research Group, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain.
| | - Peter Bauer
- Insititute of Medical Genetics and Applied Genomics, Tübingen University, Tübingen, Germany.,CENTOGENE AG, Rostock, Germany
| | | | - Thomas Klopstock
- Department of Neurology, Friedrich-Baur-Institute, University Hospital of the Ludwig-Maximilians-Universität München, Munich, Germany, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany, and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Yann Nadjar
- Department of Neurology, Reference Centre for Lysosomal Diseases (CRML), UF Neurogenetics and Metabolism, Pitié-Salpêtrière Hospital, Paris, France
| | | | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | | |
Collapse
|
84
|
Age-Specific Cut-off Values of Amino Acids and Acylcarnitines for Diagnosis of Inborn Errors of Metabolism Using Liquid Chromatography Tandem Mass Spectrometry. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3460902. [PMID: 30723736 PMCID: PMC6339774 DOI: 10.1155/2019/3460902] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 11/28/2018] [Accepted: 12/12/2018] [Indexed: 12/30/2022]
Abstract
Liquid Chromatography tandem mass spectrometry (LC-MS/MS) is used for the diagnosis of more than 30 inborn errors of metabolisms (IEMs). Accurate and reliable diagnosis of IEMs by quantifying amino acids (AAs) and acylcarnitines (ACs) using LC-MS/MS systems depend on the establishment of age-specific cut-offs of the analytes. This study aimed to (1) determine the age-specific cut-off values of AAs and ACs in Bangladesh and (2) validate the LC-MS/MS method for diagnosis of the patients with IEMs. A total of 570 enrolled healthy participants were divided into 3 age groups, namely, (1) newborns (1-7 days), (2) 8 days–7 years, and (3) 8–17 years, to establish the age-specific cut-offs for AAs and ACs. Also, 273 suspected patients with IEMs were enrolled to evaluate the reliability of the established cut-off values. Quantitation of AAs and ACs was performed on an automated LC-MS/MS system using dried blood spot (DBS) cards. Then the specimens of the enrolled clinically suspected patients were analyzed by the established method. Nine patients came out as screening positive for different IEMs, including two borderline positive cases of medium-chain acyl-CoA dehydrogenase deficiency (MCAD). A second-tier test for confirmation of the screening positive cases was conducted by urinary metabolic profiling using gas chromatography- mass spectrometry (GC-MS). Out of 9 cases that came out as screening positive by LC-MS/MS, seven cases were confirmed by urinary GC-MS analysis including 3 cases with phenylketonuria, 1 with citrullinemia type II, 1 with methylmalonic acidemia, 1 with isovaleric acidemia and 1 with carnitine uptake defect. Two borderline positive cases with MCAD were found negative by urinary GC-MS analysis. In conclusion, along with establishment of a validated LC-MS/MS method for quantitation of AAs and ACs from the DBS cards, the study also demonstrates the presence of predominantly available IEMs in Bangladesh.
Collapse
|
85
|
Abstract
Inborn errors of metabolism, also known as inherited metabolic diseases, constitute an important group of conditions presenting with neurologic signs in newborns. They are individually rare but collectively common. Many are treatable through restoration of homeostasis of a disrupted metabolic pathway. Given their frequency and potential for treatment, the clinician should be aware of this group of conditions and learn to identify the typical manifestations of the different inborn errors of metabolism. In this review, we summarize the clinical, laboratory, electrophysiologic, and neuroimaging findings of the different inborn errors of metabolism that can present with florid neurologic signs and symptoms in the neonatal period.
Collapse
MESH Headings
- Adult
- Female
- Humans
- Infant, Newborn
- Infant, Newborn, Diseases/diagnosis
- Infant, Newborn, Diseases/diagnostic imaging
- Infant, Newborn, Diseases/physiopathology
- Infant, Newborn, Diseases/therapy
- Metabolism, Inborn Errors/diagnosis
- Metabolism, Inborn Errors/diagnostic imaging
- Metabolism, Inborn Errors/physiopathology
- Metabolism, Inborn Errors/therapy
- Neuroimaging
- Pregnancy
Collapse
Affiliation(s)
- Carlos R Ferreira
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States; Rare Disease Institute, Children's National Health System, Washington, DC, United States
| | - Clara D M van Karnebeek
- Departments of Pediatrics and Clinical Genetics, Amsterdam University Medical Centers, Amsterdam, The Netherlands; Department of Pediatrics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
86
|
Carter A, Brackley SM, Gao J, Mann JP. The global prevalence and genetic spectrum of lysosomal acid lipase deficiency: A rare condition that mimics NAFLD. J Hepatol 2019; 70:142-150. [PMID: 30315827 DOI: 10.1016/j.jhep.2018.09.028] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 09/13/2018] [Accepted: 09/27/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Lysosomal acid lipase deficiency (LAL-D) is an autosomal recessive condition that may present in a mild form (cholesteryl ester storage disease [CESD]), which mimics non-alcoholic fatty liver disease (NAFLD). It has been suggested that CESD may affect 1 in 40,000 and is under-diagnosed in NAFLD clinics. Therefore, we aimed to estimate the prevalence of LAL-D using analysis of genetic variation in LIPA. METHODS MEDLINE and EMBASE were systematically searched for previously reported disease variants and prevalence estimates. Previous prevalence estimates were meta-analysed. Disease variants in LIPA were annotated with allele frequencies from gnomAD and combined with unreported major functional variants found in humans. Pooled ethnicity-specific prevalences for LAL-D and CESD were calculated using the Hardy-Weinberg equation. RESULTS Meta-analysis of existing genetic studies estimated the prevalence of LAL-D as 1 per 160,000 (95% CI 1 per 65,025-761,652) using the allele frequency of c.894G>A in LIPA. A total of 98 previously reported disease variants in LIPA were identified, of which 32/98 were present in gnomAD, giving a prevalence of 1 per 307,482 (95% CI 257,672-366,865). Wolman disease was associated with more loss-of-function variants than CESD. When this was combined with 22 previously unreported major functional variants in LIPA identified in humans, the pooled prevalence of LAL-D was 1 per 177,452 (95% CI 149,467-210,683) with a carrier frequency of 1 per 421. The prevalence is lowest in those of East Asian, South Asian, and Finnish ancestry. CONCLUSION Using 120 disease variants in LIPA, these data can reassure clinicians that LAL-D is an ultra-rare disorder. Given the therapeutic capability of sebelipase alpha, investigation for LAL-D might be included in second-line metabolic screening in NAFLD. LAY SUMMARY Lysosomal Acid Lipase Deficiency (LAL-D) is a rare genetic condition that can cause severe liver disease, but it is difficult to diagnose and sometimes can look like simple fatty liver. It was not clear how common LAL-D was and whether many cases were being missed. To study this, we searched for all genetic mutations that could cause LAL-D, calculated how common those mutations were, and added them up. This let us estimate that LAL-D affects roughly 1 in 175,000 people. We conclude that LAL-D is a very rare condition, but it is treatable so may be included in a 'second-line' of tests for causes of fatty liver.
Collapse
Affiliation(s)
- Anna Carter
- Manchester University Foundation Trust, Manchester, United Kingdom
| | - Simon Mark Brackley
- University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom
| | - Jiali Gao
- University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom
| | - Jake Peter Mann
- University of Cambridge, Department of Paediatrics, Cambridge, United Kingdom; University of Cambridge, Institute of Metabolic Science-Metabolic Research Laboratories, Cambridge, United Kingdom.
| |
Collapse
|
87
|
Pizzi MA, Alejos D, Hasan TF, Atwal PS, Krishnaiengar SR, Freeman WD. Adult Presentation of Ornithine Transcarbamylase Deficiency: 2 Illustrative Cases of Phenotypic Variability and Literature Review. Neurohospitalist 2019; 9:30-36. [PMID: 30671162 PMCID: PMC6327241 DOI: 10.1177/1941874418764817] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Ornithine transcarbamylase (OTC) deficiency is an X-linked recessive disorder that usually presents in the neonatal period. Late-onset presentation of OTC can cause mild to severe symptoms. We describe laboratory and clinical findings of late-onset presentations of OTC deficiency. We conducted a literature search using search terms "ornithine transcarbamylase deficiency," "late onset presentation," and "hyperammonemia" from January 1, 1987, to December 31, 2016, was performed. Only papers published in English were included. We searched on PubMed, MEDLINE, and Google Scholar. We also present 2 OTC deficiency cases. A total of 30 adult cases had late-onset presentation of OTC deficiency reported. The majority were women (57%) with a median age of 37 years. The median level of ammonia was 308 mmol/L and the mortality rate was 30%. Our case 1 was a 40-year-old woman who succumbed to neurologic complications after a hyperammonemia crisis following an increased protein intake. Our case 2 was a 43-year-old woman with seizures associated with increased ammonia levels. Our 2 case reports show the wide phenotypic variability and severity in late-onset presentation of OTC ranging from seizures to cerebral herniation. Our literature review is the first to detail published laboratory and neurologic sequelae of late-onset OTC deficiency.
Collapse
Affiliation(s)
| | - David Alejos
- Department of Critical Care, Mayo Clinic, Jacksonville, FL, USA
| | - Tasneem F. Hasan
- Department of Neurologic Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Paldeep S. Atwal
- Department of Clinical Genomics, Mayo Clinic, Jacksonville, FL, USA
| | | | - William D. Freeman
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
- Department of Critical Care, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
88
|
Van Mechelen K, Kessels I, Simons A, Glazemakers I. Do parents of children with metabolic diseases benefit from the Triple P - Positive Parenting Program? A pilot study. J Pediatr Endocrinol Metab 2018; 31:1335-1342. [PMID: 30433873 DOI: 10.1515/jpem-2018-0219] [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: 05/18/2018] [Accepted: 10/15/2018] [Indexed: 11/15/2022]
Abstract
Background Parents of children with metabolic diseases report more parenting stress, anxiety, depression and dysfunctional parenting styles than parents of children without metabolic diseases. In addition, their children have more behavioral problems. Beside the fact that metabolic diseases are rare, they form a relatively large proportion in the morbidity and mortality of chronically ill children. Methods In this pilot study 14 parents of children with metabolic diseases, aged between 2.5 and 13 years, participated in a quasi-experimental pre-post-follow-up study. Results After participating in the Level 4 Group Triple P-program there were small effects in decreasing child behavioral problems and large effects in decreasing dysfunctional parenting styles. There was a moderate to large reduction of parental stress and a large reduction of parental anxiety. Only the effects on the behavioral problems and the parenting style 'laxness' were no longer significant at 6 months follow-up. Conclusions In summary it can be said that the existing Triple P-program has good effects, with a great degree of satisfaction, for parents of children with metabolic diseases in reducing dysfunctional parenting styles, parenting stress and behavioral problems of their children. One should not wait for a specialized program to reach these parents, but further research is necessary as a greater effect can be expected when this program is adapted to these parents.
Collapse
Affiliation(s)
- Karen Van Mechelen
- Department of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Ilse Kessels
- Department of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - Annik Simons
- Department of Youth Mental Health, Collaborative Antwerp Psychiatric Research Institute, University of Antwerp, Wilrijk, Belgium.,Centre of Inherited Metabolic Diseases, University Hospital Antwerp, Edegem, Belgium
| | - Inge Glazemakers
- Department of Youth Mental Health, Collaborative Antwerp Psychiatric Research Institute, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
89
|
Waters D, Adeloye D, Woolham D, Wastnedge E, Patel S, Rudan I. Global birth prevalence and mortality from inborn errors of metabolism: a systematic analysis of the evidence. J Glob Health 2018; 8:021102. [PMID: 30479748 PMCID: PMC6237105 DOI: 10.7189/jogh.08.021102] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background Inborn errors of metabolism (IEM) are a group of over 500 heterogeneous disorders resulting from a defect in functioning of an intermediate metabolic pathway. Individually rare, their cumulative incidence is thought to be high, but it has not yet been estimated globally. Although outcomes can often be good if recognised early, IEM carry a high fatality rate if not diagnosed. As a result, IEM may contribute significantly to the burden of non-communicable childhood morbidity. Methods We conducted a systematic literature review of birth prevalence and case fatality of IEM globally, with search dates set from 1980 to 2017. Using random-effects meta-analysis, we estimated birth prevalence of separate classes of IEM and all-cause IEM, split by geographical region. We also estimated levels of parental consanguinity in IEM cases and global case fatality rates and resultant child deaths from all-cause IEM. Findings 49 studies met our selection criteria. We estimate the global birth prevalence of all-cause IEM to be 50.9 per 100 000 live births (95% confidence intervals (CI) = 43.4-58.4). Regional pooled birth prevalence rates showed the highest rates of IEM to be in the Eastern Mediterranean region (75.7 per 100 000 live births, 95% CI = 50.0-101.4), correlating with a higher observed rate of parental consanguinity in studies from this area. We estimate case fatality rates to be 33% or higher in low- and middle-income countries (LMICs), resulting in a minimum of 23 529 deaths from IEM per year globally (95% CI = 20 382-27 427), accounting for 0.4% of all child deaths worldwide. Conclusions IEM represent a significant cause of global child morbidity and mortality, comprising a notable proportion of child deaths currently not delineated in global modelling efforts. Our data highlight the need for policy focus on enhanced laboratory capacity for screening and diagnosis, community interventions to tackle parental consanguinity, and increased awareness and knowledge regarding management of IEM, particularly in LMICs.
Collapse
Affiliation(s)
| | | | - Daisy Woolham
- Centre for Global Health Research, The Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh Scotland, UK.,These authors contributed equally
| | - Elizabeth Wastnedge
- Centre for Global Health Research, The Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh Scotland, UK.,These authors contributed equally
| | - Smruti Patel
- Centre for Global Health Research, The Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh Scotland, UK.,These authors contributed equally
| | - Igor Rudan
- Centre for Global Health Research, The Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh Scotland, UK.,These authors contributed equally
| |
Collapse
|
90
|
Genç Sel Ç, Kılıç M, Yüksel D, Aksoy A, Kasapkara ÇS, Ceylaner S, Oğuz KK. Nonketotic hyperglycinemia: Clinical range and outcome of a rare neurometabolic disease in a single-center. Brain Dev 2018; 40:865-875. [PMID: 29929752 DOI: 10.1016/j.braindev.2018.06.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 05/26/2018] [Accepted: 06/08/2018] [Indexed: 12/01/2022]
Abstract
BACKGROUND Nonketotic hyperglycinemia (NKH) is an autosomal recessive severe life-threatening catostrophic metabolic disorder. MATERIALS AND METHODS The present study was conducted in a tertiary reference center in Turkey for six years period. The accurate diagnosis of six NKH patients was based on clinical history of the patients, neurological examinations, seizure semiology, serial electroencephalography (EEG) recordings, neuroimaging findings, metabolic tests and genetic analysis. RESULTS The common clinical findings were hypotonia with severe head lag, poor feeding, poor sucking, and intractable seizures. The starting age of the symptoms was between birth and 45 days of age (median: 8 days). The starting age of the seizures was between 30 min of age and 45 days of age (median: 18 days). The age of accurate diagnosis was between 1 month of age and 5.5 months of age (mean: 3.75 ± 1.69 months). The cerebrospinal fluid (CSF) to plasma GLY ratio of the patients was between 0.031 and 0.21 (median: 0.16). The EEG patterns of the patients were suppression-burst, hypsarrhythmia, multifocal epileptic activity, and right centro-occipital epileptic activity on admission. The neuroimaging findings were diffuse hypomyelination, corpus callosum (CC) hypoplasia, CC agenesis and brainstem hypoplasia on the magnetic resonance imaging and glycine peak was evidenced on magnetic resonance spectroscopy. Four of the patients were mutation-positive. CONCLUSIONS If a child is encephalopathic and/or hypotonic with severe head lag, early evaluation of the EEG records should be made even without a history of clinical seizures. The disease has a heterogenous course and the clinical outcome depends on the mutation type.
Collapse
Affiliation(s)
- Çiğdem Genç Sel
- Dr. Sami Ulus Pediatric and Training Hospital, Pediatric Neurology Department, Ankara, Turkey.
| | - Mustafa Kılıç
- Dr. Sami Ulus Pediatric and Training Hospital, Pediatric Metabolism Department, Ankara, Turkey
| | - Deniz Yüksel
- Dr. Sami Ulus Pediatric and Training Hospital, Pediatric Neurology Department, Ankara, Turkey
| | - Ayşe Aksoy
- Dr. Sami Ulus Pediatric and Training Hospital, Pediatric Neurology Department, Ankara, Turkey
| | - Çiğdem Seher Kasapkara
- Dr. Sami Ulus Pediatric and Training Hospital, Pediatric Metabolism Department, Ankara, Turkey
| | - Serdar Ceylaner
- Laborotory of Genetics Intergene, Genetics Department, Ankara, Turkey
| | - Kader Karlı Oğuz
- Hacettepe University of Faculty of Medicine, Radiology Department, Neuroradiology Division, Ankara, Turkey
| |
Collapse
|
91
|
Brusius-Facchin AC, Rojas Malaga D, Leistner-Segal S, Giugliani R. Recent advances in molecular testing to improve early diagnosis in children with mucopolysaccharidoses. Expert Rev Mol Diagn 2018; 18:855-866. [DOI: 10.1080/14737159.2018.1523722] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
| | - Diana Rojas Malaga
- Medical Genetics Service, HCPA, Porto Alegre, RS, Brazil
- Postgraduate Program of Genetics and Molecular Biology, UFRGS, Porto Alegre, RS, Brazil
| | - Sandra Leistner-Segal
- Medical Genetics Service, HCPA, Porto Alegre, RS, Brazil
- Postgraduate Program in Medical Science, UFRGS, Porto Alegre, RS, Brazil
| | - Roberto Giugliani
- Medical Genetics Service, HCPA, Porto Alegre, RS, Brazil
- Postgraduate Program of Genetics and Molecular Biology, UFRGS, Porto Alegre, RS, Brazil
- Postgraduate Program in Medical Science, UFRGS, Porto Alegre, RS, Brazil
- Department of Genetics, UFRGS, Porto Alegre, RS, Brazil
| |
Collapse
|
92
|
Louw R, Smuts I, Wilsenach KL, Jonck LM, Schoonen M, van der Westhuizen FH. The dilemma of diagnosing coenzyme Q 10 deficiency in muscle. Mol Genet Metab 2018. [PMID: 29530532 DOI: 10.1016/j.ymgme.2018.02.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Coenzyme Q10 (CoQ10) is an important component of the mitochondrial respiratory chain (RC) and is critical for energy production. Although the prevalence of CoQ10 deficiency is still unknown, the general consensus is that the condition is under-diagnosed. The aim of this study was to retrospectively investigate CoQ10 deficiency in frozen muscle specimens in a cohort of ethnically diverse patients who received muscle biopsies for the investigation of a possible RC deficiency (RCD). METHODS Muscle samples were homogenized whereby 600 ×g supernatants were used to analyze RC enzyme activities, followed by quantification of CoQ10 by stable isotope dilution liquid chromatography tandem mass spectrometry. The experimental group consisted of 156 patients of which 76 had enzymatically confirmed RCDs. To further assist in the diagnosis of CoQ10 deficiency in this cohort, we included sequencing of 18 selected nuclear genes involved with CoQ10 biogenesis in 26 patients with low CoQ10 concentration in muscle samples. RESULTS Central 95% reference intervals (RI) were established for CoQ10 normalized to citrate synthase (CS) or protein. Nine patients were considered CoQ10 deficient when expressed against CS, while 12 were considered deficient when expressed against protein. In two of these patients the molecular genetic cause could be confirmed, of which one would not have been identified as CoQ10 deficient if expressed only against protein content. CONCLUSION In this retrospective study, we report a central 95% reference interval for 600 ×g muscle supernatants prepared from frozen samples. The study reiterates the importance of including CoQ10 quantification as part of a diagnostic approach to study mitochondrial disease as it may complement respiratory chain enzyme assays with the possible identification of patients that may benefit from CoQ10 supplementation. However, the anomaly that only a few patients were identified as CoQ10 deficient against both markers (CS and protein), while the majority of patients where only CoQ10 deficient against one of the markers (and not the other), remains problematic. We therefore conclude from our data that, to prevent possibly not diagnosing a potential CoQ10 deficiency, the expression of CoQ10 levels in muscle on both CS as well as protein content should be considered.
Collapse
Affiliation(s)
- Roan Louw
- Human Metabolomics, North-West University (Potchefstroom Campus), Potchefstroom, South Africa.
| | - Izelle Smuts
- Department of Paediatrics and Child Health, Steve Biko Academic Hospital, University of Pretoria, Pretoria, South Africa
| | - Kimmey-Li Wilsenach
- Human Metabolomics, North-West University (Potchefstroom Campus), Potchefstroom, South Africa
| | - Lindi-Maryn Jonck
- Human Metabolomics, North-West University (Potchefstroom Campus), Potchefstroom, South Africa
| | - Maryke Schoonen
- Human Metabolomics, North-West University (Potchefstroom Campus), Potchefstroom, South Africa
| | | |
Collapse
|
93
|
Echeverri OY, Guevara JM, Espejo-Mojica ÁJ, Ardila A, Pulido N, Reyes M, Rodriguez-Lopez A, Alméciga-Díaz CJ, Barrera LA. Research, diagnosis and education in inborn errors of metabolism in Colombia: 20 years' experience from a reference center. Orphanet J Rare Dis 2018; 13:141. [PMID: 30115094 PMCID: PMC6097205 DOI: 10.1186/s13023-018-0879-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 07/26/2018] [Indexed: 12/11/2022] Open
Abstract
The use of specialized centers has been the main alternative for an appropriate diagnosis, management and follow up of patients affected by inborn errors of metabolism (IEM). These centers facilitate the training of different professionals, as well as the research at basic, translational and clinical levels. Nevertheless, few reports have described the experience of these centers and their local and/or global impact in the study of IEM. In this paper, we describe the experience of a Colombian reference center for the research, diagnosis, training and education on IEM. During the last 20 years, important advances have been achieved in the clinical knowledge of these disorders, as well as in the local availability of several diagnosis tests. Organic acidurias have been the most frequently detected diseases, followed by aminoacidopathies and peroxisomal disorders. Research efforts have been focused in the production of recombinant proteins in microorganisms towards the development of new enzyme replacement therapies, the design of gene therapy vectors and the use of bioinformatics tools for the understanding of IEM. In addition, this center has participated in the education and training of a large number professionals at different levels, which has contributed to increase the knowledge and divulgation of these disorders along the country. Noteworthy, in close collaboration with patient advocacy groups, we have participated in the discussion and construction of initiatives for the inclusion of diagnosis tests and treatments in the health system.
Collapse
Affiliation(s)
- Olga Y. Echeverri
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No 43 - 82, Building 54, Room 305A, Bogotá, Colombia
| | - Johana M. Guevara
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No 43 - 82, Building 54, Room 305A, Bogotá, Colombia
| | - Ángela J. Espejo-Mojica
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No 43 - 82, Building 54, Room 305A, Bogotá, Colombia
| | - Andrea Ardila
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No 43 - 82, Building 54, Room 305A, Bogotá, Colombia
- Clinical Laboratory – Inborn Errors of Metabolism Section, Hospital Universitario San Ignacio, Bogotá, Colombia
| | - Ninna Pulido
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No 43 - 82, Building 54, Room 305A, Bogotá, Colombia
- Clinical Laboratory – Inborn Errors of Metabolism Section, Hospital Universitario San Ignacio, Bogotá, Colombia
| | - Magda Reyes
- Clinical Laboratory – Inborn Errors of Metabolism Section, Hospital Universitario San Ignacio, Bogotá, Colombia
| | - Alexander Rodriguez-Lopez
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No 43 - 82, Building 54, Room 305A, Bogotá, Colombia
| | - Carlos J. Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No 43 - 82, Building 54, Room 305A, Bogotá, Colombia
| | - Luis A. Barrera
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No 43 - 82, Building 54, Room 305A, Bogotá, Colombia
- Clínica de Errores Innatos del Metabolismo, Hospital Universitario San Ignacio, Bogotá, Colombia
| |
Collapse
|
94
|
Lin HY, Chuang CK, Lee CL, Tu RY, Lo YT, Chiu PC, Niu DM, Fang YY, Chen TL, Tsai FJ, Hwu WL, Lin SJ, Chang TM, Lin SP. Mucopolysaccharidosis III in Taiwan: Natural history, clinical and molecular characteristics of 28 patients diagnosed during a 21-year period. Am J Med Genet A 2018; 176:1799-1809. [PMID: 30070758 DOI: 10.1002/ajmg.a.40351] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/04/2018] [Accepted: 05/07/2018] [Indexed: 01/01/2023]
Abstract
Mucopolysaccharidosis type III (MPS III, Sanfilippo syndrome) has a variable age of onset and variable rate of progression. However, information regarding the natural history of this disorder in Asian populations is limited. A retrospective analysis was carried out for 28 patients with MPS III (types IIIA [n = 3], IIIB [n = 23], and IIIC [n = 2]; 15 males and 13 females; median age, 8.2 years; age range, 2.7-26.5 years) seen in six medical centers in Taiwan from January 1996 through October 2017. The median age at confirmed diagnosis was 4.6 years. The most common initial symptom was speech delay (75%), followed by hirsutism (64%) and hyperactivity (54%). Both z scores for height and weight were negatively correlated with age (r = -.693 and -0.718, respectively; p < .01). The most prevalent clinical manifestations were speech delay (100%) and intellectual disability (100%), followed by hirsutism (93%), hyperactivity (79%), coarse facial features (68%), sleep disorders (61%), and hepatosplenomegaly (61%). Ten patients (36%) had epilepsy, and the median age at the first seizure was 11 years. Thirteen patients (46%) experienced at least one surgical procedure. At the time of the present study, 7 of the 28 patients had passed away at the median age of 13.0 years. Molecular studies showed an allelic heterogeneity without clear genotype and phenotype correlations. MPS IIIB is the most frequent subtype among MPS III in the Taiwanese population. An understanding of the natural history of MPS III may allow early diagnosis and timely management of the disease facilitating better treatment outcomes.
Collapse
Affiliation(s)
- Hsiang-Yu Lin
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan.,Department of Pediatrics, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan.,Mackay Junior College of Medicine, Nursing and Management, Taipei, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Chih-Kuang Chuang
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan.,Medical College, Fu-Jen Catholic University, Taipei, Taiwan
| | - Chung-Lin Lee
- Department of Pediatrics, Mackay Memorial Hospital, Taipei, Taiwan
| | - Ru-Yi Tu
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan
| | - Yun-Ting Lo
- Department of Laboratory Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Pao Chin Chiu
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Dau-Ming Niu
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Ya Fang
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan
| | - Tzu-Lin Chen
- Department of Laboratory Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Fuu-Jen Tsai
- Department of Pediatrics, China Medical University Hospital, Taichung, Taiwan
| | - Wuh-Liang Hwu
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Shio Jean Lin
- Department of Pediatrics, Chi Mei Medical Center, Tainan, Taiwan
| | - Tung-Ming Chang
- Department of Pediatric Neurology, Changhua Christian Children's Hospital, Changhua, Taiwan.,Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Shuan-Pei Lin
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan.,Department of Pediatrics, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Infant and Child Care, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| |
Collapse
|
95
|
van Karnebeek CDM, Wortmann SB, Tarailo-Graovac M, Langeveld M, Ferreira CR, van de Kamp JM, Hollak CE, Wasserman WW, Waterham HR, Wevers RA, Haack TB, Wanders RJA, Boycott KM. The role of the clinician in the multi-omics era: are you ready? J Inherit Metab Dis 2018; 41:571-582. [PMID: 29362952 PMCID: PMC5959952 DOI: 10.1007/s10545-017-0128-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 12/10/2017] [Accepted: 12/12/2017] [Indexed: 12/11/2022]
Abstract
Since Garrod's first description of alkaptonuria in 1902, and newborn screening for phenylketonuria introduced in the 1960s, P4 medicine (preventive, predictive, personalized, and participatory) has been a reality for the clinician serving patients with inherited metabolic diseases. The era of high-throughput technologies promises to accelerate its scale dramatically. Genomics, transcriptomics, epigenomics, proteomics, glycomics, metabolomics, and lipidomics offer an amazing opportunity for holistic investigation and contextual pathophysiologic understanding of inherited metabolic diseases for precise diagnosis and tailored treatment. While each of the -omics technologies is important to systems biology, some are more mature than others. Exome sequencing is emerging as a reimbursed test in clinics around the world, and untargeted metabolomics has the potential to serve as a single biochemical testing platform. The challenge lies in the integration and cautious interpretation of these big data, with translation into clinically meaningful information and/or action for our patients. A daunting but exciting task for the clinician; we provide clinical cases to illustrate the importance of his/her role as the connector between physicians, laboratory experts and researchers in the basic, computer, and clinical sciences. Open collaborations, data sharing, functional assays, and model organisms play a key role in the validation of -omics discoveries. Having all the right expertise at the table when discussing the diagnostic approach and individualized management plan according to the information yielded by -omics investigations (e.g., actionable mutations, novel therapeutic interventions), is the stepping stone of P4 medicine. Patient participation and the adjustment of the medical team's plan to his/her and the family's wishes most certainly is the capstone. Are you ready?
Collapse
Affiliation(s)
- Clara D M van Karnebeek
- Department of Pediatrics and Clinical Genetics, Academic Medical Centre, Amsterdam, The Netherlands.
- Departments of Pediatrics, Centre for Molecular Medicine and Therapeutics, BC Children's Research Institute, University of British Columbia, Vancouver, BC, Canada.
- Deparment of Pediatrics (Room H7-224), Emma Children's Hospital, Academic Medical Centre, Meibergdreef 9, 1105, AZ, Amsterdam, The Netherlands.
| | - Saskia B Wortmann
- Department of Pediatrics, Salzburger Landeskliniken (SALK) and Paracelsus Medical University (PMU), Salzburg, Austria
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Human Genetics, Technische Universität München, Munich, Germany
| | - Maja Tarailo-Graovac
- Departments of Pediatrics, Centre for Molecular Medicine and Therapeutics, BC Children's Research Institute, University of British Columbia, Vancouver, BC, Canada
- Departments of Medical Genetics, Centre for Molecular Medicine and Therapeutics, BC Children's Research Institute, Vancouver, BC, Canada
- Departments of Biochemistry, Molecular Biology, and Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, CA, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, CA, Canada
| | - Mirjam Langeveld
- Department of Endocrinology and Metabolism, Academic Medical Centre, Amsterdam, The Netherlands
| | - Carlos R Ferreira
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jiddeke M van de Kamp
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands
| | - Carla E Hollak
- Department of Endocrinology and Metabolism, Academic Medical Centre, Amsterdam, The Netherlands
| | - Wyeth W Wasserman
- Departments of Pediatrics, Centre for Molecular Medicine and Therapeutics, BC Children's Research Institute, University of British Columbia, Vancouver, BC, Canada
- Departments of Medical Genetics, Centre for Molecular Medicine and Therapeutics, BC Children's Research Institute, Vancouver, BC, Canada
| | - Hans R Waterham
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Laboratory Division & Department of Pediatrics, Academic Medical Centre, Amsterdam, The Netherlands
| | - Ron A Wevers
- Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Tobias B Haack
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | - Ronald J A Wanders
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Laboratory Division & Department of Pediatrics, Academic Medical Centre, Amsterdam, The Netherlands
| | - Kym M Boycott
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Canada
| |
Collapse
|
96
|
Guo K, Zhou X, Chen X, Wu Y, Liu C, Kong Q. Expanded Newborn Screening for Inborn Errors of Metabolism and Genetic Characteristics in a Chinese Population. Front Genet 2018; 9:122. [PMID: 29731766 PMCID: PMC5920142 DOI: 10.3389/fgene.2018.00122] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 03/26/2018] [Indexed: 12/24/2022] Open
Abstract
The incidence of inborn errors of metabolisms (IEMs) varies dramatically in different countries and regions. Expanded newborn screening for IEMs by tandem mass spectrometry (MS/MS) is an efficient approach for early diagnosis and presymptomatic treatment to prevent severe permanent sequelae and death. To determine the characteristics of IEMs and IEMs-associated mutations in newborns in Jining area, China, 48,297 healthy neonates were recruited for expanded newborn screening by MS/MS. The incidence of IEMs was 1/1178 in Jining, while methylmalonic acidemia, phenylketonuria, and primary carnitine deficiency ranked the top 3 of all detected IEMs. Thirty mutations in nine IEMs-associated genes were identified in 28 confirmed cases. As 19 cases with the mutations in phenylalanine hydroxylase (PAH), solute carrier family 22 member 5 (SLC22A5), and methylmalonic aciduria (cobalamin deficiency) cblC type with homocystinuria (MMACHC) genes, respectively, it suggested that mutations in the PAH, SLC22A5, and MMACHC genes are the predominant causes of IEMs, leading to the high incidence of phenylketonuria, primary carnitine deficiency, and methylmalonic acidemia, respectively. Our work indicated that the overall incidence of IEMs is high and the mutations in PAH, SLC22A5, and MMACHC genes are the leading causes of IEMs in Jining area. Therefore, it is critical to increase the coverage of expanded newborn screening by MS/MS and prenatal genetic consulting in Jining area.
Collapse
Affiliation(s)
- Kejian Guo
- Jining Maternal and Child Health Care Hospital, Jining, China
| | - Xuan Zhou
- Department of Psychiatry, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, Jining Medical University, Jining, China
| | - Xigui Chen
- Jining Maternal and Child Health Care Hospital, Jining, China
| | - Yili Wu
- Department of Psychiatry, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, Jining Medical University, Jining, China.,Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China
| | - Chuanxin Liu
- Department of Psychiatry, Jining Medical University, Jining, China.,Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China
| | - Qingsheng Kong
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China.,Department of Biochemistry, Jining Medical University, Jining, China
| |
Collapse
|
97
|
Zelei T, Csetneki K, Vokó Z, Siffel C. Epidemiology of Sanfilippo syndrome: results of a systematic literature review. Orphanet J Rare Dis 2018; 13:53. [PMID: 29631636 PMCID: PMC5891921 DOI: 10.1186/s13023-018-0796-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 03/27/2018] [Indexed: 01/30/2023] Open
Abstract
Background Sanfilippo syndrome (mucopolysaccharidosis [MPS] III subtypes A, B, C, and D) is a rare autosomal recessive inherited metabolic disorder that causes progressive neurocognitive degeneration. This systematic literature review was undertaken to compile and assess published epidemiological data, including various frequency measures and geographical variation on Sanfilippo syndrome. Methods The following databases were systematically searched for terms related to Sanfilippo syndrome epidemiology: Medline, Embase, Cochrane Database of Systematic Reviews, Academic Search Complete, Cumulative Index to Nursing and Allied Health Literature, and the Centre for Reviews and Dissemination. Qualitative synthesis of research findings was performed. Results Of 2794 publications found in the initial search, 116 were deemed eligible after title and abstract screening. Following full-text review, 46 papers were included in the qualitative synthesis. Results of this systematic literature review indicate that lifetime risk at birth ranges from 0.17–2.35 per 100,000 live births for all 4 subtypes of MPS III together, and from 0.00–1.62 per 100,000 live births for the most frequent subtype, MPS IIIA. Conclusion All 4 subtypes of MPS III are exceptionally rare, but they each have devastating effects on children. Higher-quality epidemiological data are needed to appropriately target resources for disease research and management. Electronic supplementary material The online version of this article (10.1186/s13023-018-0796-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tamás Zelei
- Syreon Research Institute, Budapest, Hungary
| | | | - Zoltán Vokó
- Syreon Research Institute, Budapest, Hungary
| | - Csaba Siffel
- Shire, 300 Shire Way, Lexington, MA, 02421, USA.
| |
Collapse
|
98
|
Borger DK, McMahon B, Roshan Lal T, Serra-Vinardell J, Aflaki E, Sidransky E. Induced pluripotent stem cell models of lysosomal storage disorders. Dis Model Mech 2018; 10:691-704. [PMID: 28592657 PMCID: PMC5483008 DOI: 10.1242/dmm.029009] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 04/28/2017] [Indexed: 01/30/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) have provided new opportunities to explore the cell biology and pathophysiology of human diseases, and the lysosomal storage disorder research community has been quick to adopt this technology. Patient-derived iPSC models have been generated for a number of lysosomal storage disorders, including Gaucher disease, Pompe disease, Fabry disease, metachromatic leukodystrophy, the neuronal ceroid lipofuscinoses, Niemann-Pick types A and C1, and several of the mucopolysaccharidoses. Here, we review the strategies employed for reprogramming and differentiation, as well as insights into disease etiology gleaned from the currently available models. Examples are provided to illustrate how iPSC-derived models can be employed to develop new therapeutic strategies for these disorders. We also discuss how models of these rare diseases could contribute to an enhanced understanding of more common neurodegenerative disorders such as Parkinson’s disease, and discuss key challenges and opportunities in this area of research. Summary: This Review discusses how induced pluripotent stem cells (iPSCs) provide new opportunities to explore the biology and pathophysiology of lysosomal storage diseases, and how iPSCs have illuminated the role of lysosomes in more common disorders.
Collapse
Affiliation(s)
- Daniel K Borger
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Benjamin McMahon
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tamanna Roshan Lal
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jenny Serra-Vinardell
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elma Aflaki
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ellen Sidransky
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
99
|
Laboratory analysis of organic acids, 2018 update: a technical standard of the American College of Medical Genetics and Genomics (ACMG). Genet Med 2018. [DOI: 10.1038/gim.2018.45] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
100
|
Hannah-Shmouni F, Stratakis CA. An overview of inborn errors of metabolism manifesting with primary adrenal insufficiency. Rev Endocr Metab Disord 2018; 19:53-67. [PMID: 29956047 PMCID: PMC6204320 DOI: 10.1007/s11154-018-9447-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Primary adrenal insufficiency (PAI) results from an inability to produce adequate amounts of steroid hormones from the adrenal cortex. The most common causes of PAI are autoimmune adrenalitis (Addison's disease), infectious diseases, adrenalectomy, neoplasia, medications, and various rare genetic syndromes and inborn errors of metabolism that typically present in childhood although late-onset presentations are becoming increasingly recognized. The prevalence of PAI in Western countries is approximately 140 cases per million, with an incidence of 4 per 1,000,000 per year. Several pitfalls in the genetic diagnosis of patients with PAI exist. In this review, we provide an in-depth discussion and overview on the inborn errors of metabolism manifesting with PAI, including genetic diagnosis, genotype-phenotype relationships and counseling of patients and their families with a focus on various enzymatic deficiencies of steroidogenesis.
Collapse
Affiliation(s)
- Fady Hannah-Shmouni
- Section on Endocrinology & Genetics, The Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Building 10, CRC, Room 1-3330, 10 Center Dr., MSC1103, Bethesda, MD, 20892, USA
| | - Constantine A Stratakis
- Section on Endocrinology & Genetics, The Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Building 10, CRC, Room 1-3330, 10 Center Dr., MSC1103, Bethesda, MD, 20892, USA.
| |
Collapse
|