1
|
Beltran-Velasco AI. Brain Glycogen-Its Metabolic Role in Neuronal Health and Neurological Disorders-An Extensive Narrative Review. Metabolites 2025; 15:128. [PMID: 39997753 PMCID: PMC11857135 DOI: 10.3390/metabo15020128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/06/2025] [Revised: 01/29/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
Background: Brain glycogen is imperative for neuronal health, as it supports energy demands and metabolic processes. This review examines the pathways involved in glycogen storage and utilization in the central nervous system, emphasizing their role in both physiology and pathology. It explores how alterations in glycogen metabolism contribute to neurological disorders, including neurodegenerative diseases, epilepsy, and metabolic conditions while highlighting the bidirectional interaction between neurons and glia in maintaining brain homeostasis. Methods: A comprehensive search of articles published between 2015 and 2025 was conducted using the following databases: ScienceDirect, Scopus, Wiley, Web of Science, Medline, and PubMed. The selection of relevant studies was based on their focus on brain glycogen metabolism and its role in neurological conditions, with studies that did not meet the inclusion criteria being excluded. Results: The metabolic processes of brain glycogen are subject to rigorous regulation by astrocyte-neuron interactions, thereby ensuring metabolic homeostasis and energy availability. The dysregulation of glycogen storage and mobilization has been implicated in the development of synaptic dysfunction, excitotoxicity, and neurodegeneration in a variety of disorders. For instance, aberrant glycogen accumulation in diseases such as Lafora disease has been associated with severe neurodegeneration, while impaired glycogen mobilization has been shown to exacerbate energy deficits in Alzheimer's and epilepsy. Conclusions: Targeting brain glycogen metabolism represents a promising approach for therapeutic intervention in neurological disorders. However, the translation of these strategies to human models remains challenging, particularly with regard to the long-term safety and specificity of glycogen-targeted therapies.
Collapse
|
2
|
Agnoletto L, Vandeleur M, White M, Adams A, Halligan R, Peters H. Sleep quality in children with hepatic glycogen storage diseases, a prospective observational pilot study. JIMD Rep 2025; 66:e12462. [PMID: 39723121 PMCID: PMC11667772 DOI: 10.1002/jmd2.12462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 07/24/2024] [Revised: 10/16/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Background Hepatic glycogen storage diseases (GSDs) are characterised by enzyme defects affecting liver glycogen metabolism, where carbohydrate supplementation to prevent overnight hypoglycaemia is common. Concerns around sleep quality in hepatic GSDs relate to emerging evidence that overnight dysglycaemia impacts sleep quality. Methods This prospective observational study reported sleep quality and duration in children with hepatic GSDs over 7 days utilising: actigraphy (Actiwatch 2 by Phillips Respironics), sleep diaries, proxy reported age-appropriate sleep and quality-of-life (QoL) questionnaires, in the context of nocturnal glycaemic profiles continuous glucose monitor (CGM, Dexcom G6) and nocturnal dietary management strategies. Significant hypo- and hyperglycaemia were defined as ≥1% of sleep diary documented nocturnal period, recording <3.5 and >10.0 mmol/L, respectively. Results Seven children with hepatic GSD (aged 1-17 years) participated. Objective sleep quality was poor, with actigraphy demonstrating that no child achieved the minimum sleep duration recommended for age. Subjective sleep quality was also poor, with 4/5 documenting significant daytime sleepiness and 6/6 reporting poor sleep hygiene. Children prescribed overnight bolus feeds (OBF) (n = 2) recorded shorter sleep duration compared to other nocturnal management strategies. Parent-reported QoL suggested poor disease-related QoL outcomes for this cohort. Conclusion Objective and subjective sleep disturbances and reduced QoL are common within our sample of children with hepatic GSD. From our observations these outcomes may be linked to nutritional overnight interventions, especially OBFs, rather than overnight glucose levels. Consideration of the impacts of overnight feeding strategies on sleep quality and QoL in children with hepatic GSD should inform future management strategies.
Collapse
Affiliation(s)
- Lucas Agnoletto
- Murdoch Children's Research InstituteMelbourneVictoriaAustralia
- Department of PaediatricsUniversity of MelbourneMelbourneVictoriaAustralia
| | - Moya Vandeleur
- Murdoch Children's Research InstituteMelbourneVictoriaAustralia
- Department of Respiratory and Sleep MedicineRoyal Children's HospitalMelbourneVictoriaAustralia
| | - Mary White
- Murdoch Children's Research InstituteMelbourneVictoriaAustralia
- Department of Endocrinology and DiabetesRoyal Children's HospitalMelbourneVictoriaAustralia
- Melbourne School of Global and Population Health, University of MelbourneMelbourneVictoriaAustralia
| | - Anne‐Marie Adams
- Murdoch Children's Research InstituteMelbourneVictoriaAustralia
- Department of PaediatricsUniversity of MelbourneMelbourneVictoriaAustralia
- Department of Respiratory and Sleep MedicineRoyal Children's HospitalMelbourneVictoriaAustralia
| | - Rebecca Halligan
- Department of Metabolic MedicineRoyal Children's HospitalMelbourneVictoriaAustralia
- Department of Inherited Metabolic DiseasesEvelina London Children's HospitalLondonUK
| | - Heidi Peters
- Murdoch Children's Research InstituteMelbourneVictoriaAustralia
- Department of PaediatricsUniversity of MelbourneMelbourneVictoriaAustralia
- Department of Metabolic MedicineRoyal Children's HospitalMelbourneVictoriaAustralia
| |
Collapse
|
3
|
Kolbe AB, Acord MR, Khanna G, Morin CE, Nguyen HN, Rees MA, Ro E, Schooler GR, Squires JH, Syed AB, Tang ER, Towbin AJ, Alazraki A. Imaging Findings and Management Strategies for Liver Masses in Children with Predisposition Disorders: A Review by the Pediatric LI-RADS Group. Radiographics 2025; 45:e240063. [PMID: 39666572 DOI: 10.1148/rg.240063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/14/2024]
Abstract
Liver masses in children with underlying systemic disease or a predisposing syndrome can be benign or malignant, ranging from focal fat to hepatocellular carcinoma (HCC). Knowledge of the underlying condition, the pathophysiologic effect on the liver, and the development of liver disease and specific liver lesions allows radiologists to guide imaging with regard to modality and frequency and give recommendations for biopsy when appropriate. In some predisposition disorders, such as Beckwith Wiedemann spectrum, familial adenomatous polyposis syndrome, and tuberous sclerosis complex, established guidelines for imaging screening exist. In many of the syndromes discussed, masses may occur outside of the liver and the liver may not be the primary focus of screening. For other entities, no consensus recommendations exist. Screening recommendations may be based on the risk of development of chronic liver disease. Once cirrhosis occurs, the risk of developing HCC is elevated. The authors summarize the spectrum of liver lesions that may be encountered in children with predisposing syndromes and systemic diseases, the imaging appearance of the lesions with various modalities, and screening guidelines where published. ©RSNA, 2024 See the invited commentary by Rutten and Chavan in this issue.
Collapse
Affiliation(s)
- Amy B Kolbe
- From the Department of Radiology, Mayo Clinic, 200 1st Ave SE, Rochester, MN 55905 (A.B.K.); Department of Radiology, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pa (M.R.A.); Department of Radiology and Imaging Sciences, Emory University and Children's Healthcare of Atlanta, Atlanta, Ga (G.K., A.A.); Department of Radiology, Cincinnati Children's Hospital, Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio (C.E.M., A.J.T.); Department of Radiology, Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, Calif (H.N.N.); Department of Radiology, Nationwide Children's Hospital, Columbus, Ohio (M.A.R.); Department of Medical Imaging, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill (E.R.); Department of Radiology, UT Southwestern Medical Center, Dallas, Tex (G.R.S.); Department of Radiology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa (J.H.S.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.B.S.); and Department of Radiology, Children's Hospital Colorado, Aurora, Colo (E.R.T.)
| | - Michael R Acord
- From the Department of Radiology, Mayo Clinic, 200 1st Ave SE, Rochester, MN 55905 (A.B.K.); Department of Radiology, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pa (M.R.A.); Department of Radiology and Imaging Sciences, Emory University and Children's Healthcare of Atlanta, Atlanta, Ga (G.K., A.A.); Department of Radiology, Cincinnati Children's Hospital, Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio (C.E.M., A.J.T.); Department of Radiology, Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, Calif (H.N.N.); Department of Radiology, Nationwide Children's Hospital, Columbus, Ohio (M.A.R.); Department of Medical Imaging, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill (E.R.); Department of Radiology, UT Southwestern Medical Center, Dallas, Tex (G.R.S.); Department of Radiology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa (J.H.S.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.B.S.); and Department of Radiology, Children's Hospital Colorado, Aurora, Colo (E.R.T.)
| | - Geetika Khanna
- From the Department of Radiology, Mayo Clinic, 200 1st Ave SE, Rochester, MN 55905 (A.B.K.); Department of Radiology, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pa (M.R.A.); Department of Radiology and Imaging Sciences, Emory University and Children's Healthcare of Atlanta, Atlanta, Ga (G.K., A.A.); Department of Radiology, Cincinnati Children's Hospital, Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio (C.E.M., A.J.T.); Department of Radiology, Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, Calif (H.N.N.); Department of Radiology, Nationwide Children's Hospital, Columbus, Ohio (M.A.R.); Department of Medical Imaging, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill (E.R.); Department of Radiology, UT Southwestern Medical Center, Dallas, Tex (G.R.S.); Department of Radiology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa (J.H.S.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.B.S.); and Department of Radiology, Children's Hospital Colorado, Aurora, Colo (E.R.T.)
| | - Cara E Morin
- From the Department of Radiology, Mayo Clinic, 200 1st Ave SE, Rochester, MN 55905 (A.B.K.); Department of Radiology, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pa (M.R.A.); Department of Radiology and Imaging Sciences, Emory University and Children's Healthcare of Atlanta, Atlanta, Ga (G.K., A.A.); Department of Radiology, Cincinnati Children's Hospital, Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio (C.E.M., A.J.T.); Department of Radiology, Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, Calif (H.N.N.); Department of Radiology, Nationwide Children's Hospital, Columbus, Ohio (M.A.R.); Department of Medical Imaging, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill (E.R.); Department of Radiology, UT Southwestern Medical Center, Dallas, Tex (G.R.S.); Department of Radiology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa (J.H.S.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.B.S.); and Department of Radiology, Children's Hospital Colorado, Aurora, Colo (E.R.T.)
| | - HaiThuy N Nguyen
- From the Department of Radiology, Mayo Clinic, 200 1st Ave SE, Rochester, MN 55905 (A.B.K.); Department of Radiology, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pa (M.R.A.); Department of Radiology and Imaging Sciences, Emory University and Children's Healthcare of Atlanta, Atlanta, Ga (G.K., A.A.); Department of Radiology, Cincinnati Children's Hospital, Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio (C.E.M., A.J.T.); Department of Radiology, Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, Calif (H.N.N.); Department of Radiology, Nationwide Children's Hospital, Columbus, Ohio (M.A.R.); Department of Medical Imaging, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill (E.R.); Department of Radiology, UT Southwestern Medical Center, Dallas, Tex (G.R.S.); Department of Radiology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa (J.H.S.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.B.S.); and Department of Radiology, Children's Hospital Colorado, Aurora, Colo (E.R.T.)
| | - Mitchell A Rees
- From the Department of Radiology, Mayo Clinic, 200 1st Ave SE, Rochester, MN 55905 (A.B.K.); Department of Radiology, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pa (M.R.A.); Department of Radiology and Imaging Sciences, Emory University and Children's Healthcare of Atlanta, Atlanta, Ga (G.K., A.A.); Department of Radiology, Cincinnati Children's Hospital, Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio (C.E.M., A.J.T.); Department of Radiology, Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, Calif (H.N.N.); Department of Radiology, Nationwide Children's Hospital, Columbus, Ohio (M.A.R.); Department of Medical Imaging, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill (E.R.); Department of Radiology, UT Southwestern Medical Center, Dallas, Tex (G.R.S.); Department of Radiology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa (J.H.S.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.B.S.); and Department of Radiology, Children's Hospital Colorado, Aurora, Colo (E.R.T.)
| | - Esther Ro
- From the Department of Radiology, Mayo Clinic, 200 1st Ave SE, Rochester, MN 55905 (A.B.K.); Department of Radiology, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pa (M.R.A.); Department of Radiology and Imaging Sciences, Emory University and Children's Healthcare of Atlanta, Atlanta, Ga (G.K., A.A.); Department of Radiology, Cincinnati Children's Hospital, Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio (C.E.M., A.J.T.); Department of Radiology, Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, Calif (H.N.N.); Department of Radiology, Nationwide Children's Hospital, Columbus, Ohio (M.A.R.); Department of Medical Imaging, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill (E.R.); Department of Radiology, UT Southwestern Medical Center, Dallas, Tex (G.R.S.); Department of Radiology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa (J.H.S.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.B.S.); and Department of Radiology, Children's Hospital Colorado, Aurora, Colo (E.R.T.)
| | - Gary R Schooler
- From the Department of Radiology, Mayo Clinic, 200 1st Ave SE, Rochester, MN 55905 (A.B.K.); Department of Radiology, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pa (M.R.A.); Department of Radiology and Imaging Sciences, Emory University and Children's Healthcare of Atlanta, Atlanta, Ga (G.K., A.A.); Department of Radiology, Cincinnati Children's Hospital, Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio (C.E.M., A.J.T.); Department of Radiology, Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, Calif (H.N.N.); Department of Radiology, Nationwide Children's Hospital, Columbus, Ohio (M.A.R.); Department of Medical Imaging, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill (E.R.); Department of Radiology, UT Southwestern Medical Center, Dallas, Tex (G.R.S.); Department of Radiology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa (J.H.S.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.B.S.); and Department of Radiology, Children's Hospital Colorado, Aurora, Colo (E.R.T.)
| | - Judy H Squires
- From the Department of Radiology, Mayo Clinic, 200 1st Ave SE, Rochester, MN 55905 (A.B.K.); Department of Radiology, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pa (M.R.A.); Department of Radiology and Imaging Sciences, Emory University and Children's Healthcare of Atlanta, Atlanta, Ga (G.K., A.A.); Department of Radiology, Cincinnati Children's Hospital, Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio (C.E.M., A.J.T.); Department of Radiology, Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, Calif (H.N.N.); Department of Radiology, Nationwide Children's Hospital, Columbus, Ohio (M.A.R.); Department of Medical Imaging, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill (E.R.); Department of Radiology, UT Southwestern Medical Center, Dallas, Tex (G.R.S.); Department of Radiology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa (J.H.S.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.B.S.); and Department of Radiology, Children's Hospital Colorado, Aurora, Colo (E.R.T.)
| | - Ali B Syed
- From the Department of Radiology, Mayo Clinic, 200 1st Ave SE, Rochester, MN 55905 (A.B.K.); Department of Radiology, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pa (M.R.A.); Department of Radiology and Imaging Sciences, Emory University and Children's Healthcare of Atlanta, Atlanta, Ga (G.K., A.A.); Department of Radiology, Cincinnati Children's Hospital, Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio (C.E.M., A.J.T.); Department of Radiology, Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, Calif (H.N.N.); Department of Radiology, Nationwide Children's Hospital, Columbus, Ohio (M.A.R.); Department of Medical Imaging, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill (E.R.); Department of Radiology, UT Southwestern Medical Center, Dallas, Tex (G.R.S.); Department of Radiology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa (J.H.S.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.B.S.); and Department of Radiology, Children's Hospital Colorado, Aurora, Colo (E.R.T.)
| | - Elizabeth R Tang
- From the Department of Radiology, Mayo Clinic, 200 1st Ave SE, Rochester, MN 55905 (A.B.K.); Department of Radiology, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pa (M.R.A.); Department of Radiology and Imaging Sciences, Emory University and Children's Healthcare of Atlanta, Atlanta, Ga (G.K., A.A.); Department of Radiology, Cincinnati Children's Hospital, Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio (C.E.M., A.J.T.); Department of Radiology, Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, Calif (H.N.N.); Department of Radiology, Nationwide Children's Hospital, Columbus, Ohio (M.A.R.); Department of Medical Imaging, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill (E.R.); Department of Radiology, UT Southwestern Medical Center, Dallas, Tex (G.R.S.); Department of Radiology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa (J.H.S.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.B.S.); and Department of Radiology, Children's Hospital Colorado, Aurora, Colo (E.R.T.)
| | - Alexander J Towbin
- From the Department of Radiology, Mayo Clinic, 200 1st Ave SE, Rochester, MN 55905 (A.B.K.); Department of Radiology, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pa (M.R.A.); Department of Radiology and Imaging Sciences, Emory University and Children's Healthcare of Atlanta, Atlanta, Ga (G.K., A.A.); Department of Radiology, Cincinnati Children's Hospital, Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio (C.E.M., A.J.T.); Department of Radiology, Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, Calif (H.N.N.); Department of Radiology, Nationwide Children's Hospital, Columbus, Ohio (M.A.R.); Department of Medical Imaging, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill (E.R.); Department of Radiology, UT Southwestern Medical Center, Dallas, Tex (G.R.S.); Department of Radiology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa (J.H.S.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.B.S.); and Department of Radiology, Children's Hospital Colorado, Aurora, Colo (E.R.T.)
| | - Adina Alazraki
- From the Department of Radiology, Mayo Clinic, 200 1st Ave SE, Rochester, MN 55905 (A.B.K.); Department of Radiology, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pa (M.R.A.); Department of Radiology and Imaging Sciences, Emory University and Children's Healthcare of Atlanta, Atlanta, Ga (G.K., A.A.); Department of Radiology, Cincinnati Children's Hospital, Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio (C.E.M., A.J.T.); Department of Radiology, Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, Calif (H.N.N.); Department of Radiology, Nationwide Children's Hospital, Columbus, Ohio (M.A.R.); Department of Medical Imaging, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill (E.R.); Department of Radiology, UT Southwestern Medical Center, Dallas, Tex (G.R.S.); Department of Radiology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa (J.H.S.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.B.S.); and Department of Radiology, Children's Hospital Colorado, Aurora, Colo (E.R.T.)
| |
Collapse
|
4
|
Geramizadeh B, Ezgu F, Beyzaei Z. Glycogen storage disorder types IX: the mutation spectrum and ethnic distribution. Orphanet J Rare Dis 2024; 19:475. [PMID: 39707443 DOI: 10.1186/s13023-024-03488-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/25/2024] [Accepted: 11/29/2024] [Indexed: 12/23/2024] Open
Abstract
Glycogen storage disorders (GSD) GSD-IX are characterized by deficiencies in muscular and/or hepatic phosphorylase enzymes. GSD type IX za is an X-linked disorder, while IXb and IXc are autosomal recessive disorders resulting from pathogenic variants in the genes encoding the Phosphorylase b Kinase regulatory subunit alpha (PHKA), beta (PHKB), and gamma (PHKG), respectively. Despite progress in understanding these diseases, there are still unclear questions regarding their clinical manifestations, genetic variations, and the relationship between genotype and phenotype. Therefore, this review focuses on variants of GSD IX subtypes and all clinical findings to establish a genotype-phenotype relationship as well as highlighting the wide spectrum of disease-causing variants. Such information is beneficial for the establishment of a privileged mutation screening process in a specific region or ethnic group. Diagnosis is based on clinical manifestations and laboratory test results, but molecular analysis is often necessary to distinguish the various forms with similar presentations.
Collapse
Affiliation(s)
- Bita Geramizadeh
- Department of Pathology, Medical School of Shiraz University, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatih Ezgu
- Department of Pediatric Metabolism and Genetics, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Zahra Beyzaei
- Assistant Professor of Cellular and Molecular Medicine, Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Khalili St., Research Tower, Seventh Floor, Shiraz, Iran.
| |
Collapse
|
5
|
Vanduangden J, Ittiwut R, Ittiwut C, Phewplung T, Sanpavat A, Sintusek P, Suphapeetiporn K. Molecular profiles and long-term outcomes of Thai children with hepatic glycogen storage disease in Thailand. World J Clin Pediatr 2024; 13:100493. [PMID: 39654668 PMCID: PMC11572614 DOI: 10.5409/wjcp.v13.i4.100493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 08/18/2024] [Revised: 09/28/2024] [Accepted: 10/16/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Thus far, genetic analysis of patients clinically diagnosed with glycogen storage diseases (GSDs) in Thailand has not been reported. AIM To evaluate the clinical and biochemical profiles, molecular analysis and long-term outcomes of Thai children diagnosed with hepatic GSD. METHODS Children aged < 18 years diagnosed with hepatic GSD and followed up at King Chulalongkorn Memorial Hospital were recruited. Whole-exome sequencing (WES) was performed to identify the causative gene variants. Medical records were assessed. RESULTS All eight children with histopathologically confirmed diagnosis were classified by WES into subtypes Ia (n = 1), III (n = 3), VI (n = 3), and IX (n = 1). A total number of 10 variants were identified including G6PC (n = 1), AGL (n = 4), PYGL (n = 5), and PHKA2 (n = 1). AGL had two novel variants. The clinical manifestations were hepatomegaly (n = 8), doll-like facies (n = 3), wasting (n = 2), and stunting (n = 5). All patients showed hypoglycemia, transaminitis, and dyslipidemia. The mainstay of treatment was cornstarch supplementation and high-protein and low-lactose-fructose diet. After a median follow-up time of 9.59 years, height turned to normal for age in 3/5 patients and none had malnutrition. Liver enzymes, blood sugar, and lipid profiles improved in all. CONCLUSION Hepatomegaly, transaminitis, and hypoglycemia are the hallmarks of GSD confirmed by liver histopathology. Molecular analysis can confirm the diagnosis or classify the subtype that might benefit from personalized treatment, prognosis, and long-term care.
Collapse
Affiliation(s)
| | - Rungnapa Ittiwut
- Center of Excellence for Medical Genomics, Department of Pediatrics, Excellence Center for Genomics and Precision Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Chupong Ittiwut
- Department of Pediatrics, Excellence Center of Genomics and Precision Medicine, Bangkok 10330, Thailand
| | - Teerasak Phewplung
- Department of Radiology, Chulalongkorn University, Bangkok 10330, Thailand
| | - Anapat Sanpavat
- Department of Pathology, Chulalongkorn University, Bangkok 10330, Thailand
| | - Palittiya Sintusek
- Center of Excellence in Thai Pediatric Gastroenterology, Hepatology and Immunology, Division of Gastroenterology, Department of Pediatrics, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kanya Suphapeetiporn
- Center of Excellence for Medical Genetics, Department of Pediatrics, King Chulalongkorn Mem Hosp, Dept Pediat, Div Med Genet and Metab, Sor Kor Bldg, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
6
|
Gibson RA, Jeck WR, Koch RL, Mehta A, Choi SJ, Sriraman Y, Bali D, Young S, Asokan A, Lim JA, Kishnani PS. Progressive liver disease and dysregulated glycogen metabolism in murine GSD IX γ2 models human disease. Mol Genet Metab 2024; 143:108597. [PMID: 39488079 PMCID: PMC11633833 DOI: 10.1016/j.ymgme.2024.108597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 10/23/2024] [Accepted: 10/26/2024] [Indexed: 11/04/2024]
Abstract
Hepatic glycogen storage disease type IX γ2 (GSD IX γ2) is a severe, liver-specific subtype of GSD IX. While all patients with hepatic GSD IX present with similar symptoms, over 95 % of patients with GSD IX γ2 progress to liver fibrosis and cirrhosis. Despite disease severity, the long-term natural history of GSD IX γ2 liver disease progression is not known. Our lab previously characterized the Phkg2-/- mouse model at 3 months of age, demonstrating that the mouse recapitulates the early liver disease phenotype of GSD IX γ2. To understand how liver disease progresses in GSD IX γ2, we characterized the mouse model through 24 months of age. Our study showed for the first time that GSD IX γ2 mice develop liver fibrosis that progresses to cirrhosis. Importantly, we observed that the progression of liver fibrosis is associated with an initial elevation and subsequent decrease of key GSD biomarkers - the latter being a finding that is often considered to be an improvement of disease in patients. In recognition of the unique liver fibrosis pattern and to support future therapeutic investigations using this model, we developed a novel scoring system for GSD IX γ2 mouse liver pathology. Lastly, this work introduces evidence of a dysregulated glycogen metabolism pathway which can serve as an endpoint for future therapeutic evaluation. As we await longitudinal clinical natural history data, these findings greatly expand our understanding of liver disease manifestations in GSD IX γ2 and have notable clinical applications.
Collapse
Affiliation(s)
- Rebecca A Gibson
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA; Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - William R Jeck
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Rebecca L Koch
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Aarav Mehta
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Su Jin Choi
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Yajur Sriraman
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Deeksha Bali
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Sarah Young
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Aravind Asokan
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA; Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Jeong-A Lim
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Priya S Kishnani
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA; Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
7
|
Overduin RJ, Venema A, Lubout CMA, Fokkert-Wilts MJ, De Boer F, Schreuder AB, Rossi A, Derks TGJ. Continuous glucose monitoring metrics in people with liver glycogen storage disease and idiopathic ketotic hypoglycemia: A single-center, retrospective, observational study. Mol Genet Metab 2024; 143:108573. [PMID: 39243574 DOI: 10.1016/j.ymgme.2024.108573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 06/28/2024] [Revised: 08/20/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Cohort data on continuous glucose monitoring (CGM) metrics are scarce for liver glycogen storage diseases (GSDs) and idiopathic ketotic hypoglycemia (IKH). The aim of this study was to retrospectively describe CGM metrics for people with liver GSDs and IKH. PATIENTS AND METHODS CGM metrics (descriptive, glycemic variation and glycemic control parameters) were calculated for 47 liver GSD and 14 IKH patients, categorized in cohorts by disease subtype, age and treatment status, and compared to published age-matched CGM metrics from healthy individuals. Glycemic control was assessed as time-in-range (TIR; ≥3.9 - ≤7.8 and ≥3.9 - ≤10.0 mmol/L), time-below-range (TBR; <3.0 mmol/L and ≥3.0 - ≤3.9 mmol/L), and time-above-range (TAR; >7.8 and >10.0 mmol/L). RESULTS Despite all patients receiving dietary treatment, GSD cohorts displayed significantly different CGM metrics compared to healthy individuals. Decreased TIR together with increased TAR were noted in GSD I, GSD III, and GSD XI (Fanconi-Bickel syndrome) cohorts (all p < 0.05). In addition, all GSD I cohorts showed increased TBR (all p < 0.05). In GSD IV an increased TBR (p < 0.05) and decreased TAR were noted (p < 0.05). In GSD IX only increased TAR was observed (p < 0.05). IKH patient cohorts, both with and without treatment, presented CGM metrics similar to healthy individuals. CONCLUSION Despite dietary treatment, most liver GSD cohorts do not achieve CGM metrics comparable to healthy individuals. International recommendations on the use of CGM and clinical targets for CGM metrics in liver GSD patients are warranted, both for patient care and clinical trials.
Collapse
Affiliation(s)
- Ruben J Overduin
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Annieke Venema
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Charlotte M A Lubout
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marieke J Fokkert-Wilts
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Foekje De Boer
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Andrea B Schreuder
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Alessandro Rossi
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Translational Medicine, Section of Pediatrics, University of Naples "Federico II", Naples, Italy
| | - Terry G J Derks
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
8
|
Mishra K, Kakhlon O. Mitochondrial Dysfunction in Glycogen Storage Disorders (GSDs). Biomolecules 2024; 14:1096. [PMID: 39334863 PMCID: PMC11430448 DOI: 10.3390/biom14091096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/30/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
Glycogen storage disorders (GSDs) are a group of inherited metabolic disorders characterized by defects in enzymes involved in glycogen metabolism. Deficiencies in enzymes responsible for glycogen breakdown and synthesis can impair mitochondrial function. For instance, in GSD type II (Pompe disease), acid alpha-glucosidase deficiency leads to lysosomal glycogen accumulation, which secondarily impacts mitochondrial function through dysfunctional mitophagy, which disrupts mitochondrial quality control, generating oxidative stress. In GSD type III (Cori disease), the lack of the debranching enzyme causes glycogen accumulation and affects mitochondrial dynamics and biogenesis by disrupting the integrity of muscle fibers. Malfunctional glycogen metabolism can disrupt various cascades, thus causing mitochondrial and cell metabolic dysfunction through various mechanisms. These dysfunctions include altered mitochondrial morphology, impaired oxidative phosphorylation, increased production of reactive oxygen species (ROS), and defective mitophagy. The oxidative burden typical of GSDs compromises mitochondrial integrity and exacerbates the metabolic derangements observed in GSDs. The intertwining of mitochondrial dysfunction and GSDs underscores the complexity of these disorders and has significant clinical implications. GSD patients often present with multisystem manifestations, including hepatomegaly, hypoglycemia, and muscle weakness, which can be exacerbated by mitochondrial impairment. Moreover, mitochondrial dysfunction may contribute to the progression of GSD-related complications, such as cardiomyopathy and neurocognitive deficits. Targeting mitochondrial dysfunction thus represents a promising therapeutic avenue in GSDs. Potential strategies include antioxidants to mitigate oxidative stress, compounds that enhance mitochondrial biogenesis, and gene therapy to correct the underlying mitochondrial enzyme deficiencies. Mitochondrial dysfunction plays a critical role in the pathophysiology of GSDs. Recognizing and addressing this aspect can lead to more comprehensive and effective treatments, improving the quality of life of GSD patients. This review aims to elaborate on the intricate relationship between mitochondrial dysfunction and various types of GSDs. The review presents challenges and treatment options for several GSDs.
Collapse
Affiliation(s)
- Kumudesh Mishra
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 9112001, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Ein Kerem, Jerusalem 9112102, Israel
| | - Or Kakhlon
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 9112001, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Ein Kerem, Jerusalem 9112102, Israel
| |
Collapse
|
9
|
Rossi A, Simeoli C, Pivonello R, Salerno M, Rosano C, Brunetti B, Strisciuglio P, Colao A, Parenti G, Melis D, Derks TGJ. Endocrine involvement in hepatic glycogen storage diseases: pathophysiology and implications for care. Rev Endocr Metab Disord 2024; 25:707-725. [PMID: 38556561 PMCID: PMC11294274 DOI: 10.1007/s11154-024-09880-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Accepted: 03/23/2024] [Indexed: 04/02/2024]
Abstract
Hepatic glycogen storage diseases constitute a group of disorders due to defects in the enzymes and transporters involved in glycogen breakdown and synthesis in the liver. Although hypoglycemia and hepatomegaly are the primary manifestations of (most of) hepatic GSDs, involvement of the endocrine system has been reported at multiple levels in individuals with hepatic GSDs. While some endocrine abnormalities (e.g., hypothalamic‑pituitary axis dysfunction in GSD I) can be direct consequence of the genetic defect itself, others (e.g., osteopenia in GSD Ib, insulin-resistance in GSD I and GSD III) may be triggered by the (dietary/medical) treatment. Being aware of the endocrine abnormalities occurring in hepatic GSDs is essential (1) to provide optimized medical care to this group of individuals and (2) to drive research aiming at understanding the disease pathophysiology. In this review, a thorough description of the endocrine manifestations in individuals with hepatic GSDs is presented, including pathophysiological and clinical implications.
Collapse
Affiliation(s)
- Alessandro Rossi
- Section of Metabolic Diseases, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
- Department of Translational Medicine, Section of Pediatrics, University of Naples "Federico II", Naples, Italy.
| | - Chiara Simeoli
- Dipartmento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Diabetologia ed Andrologia, University of Naples "Federico II", Naples, Italy
| | - Rosario Pivonello
- Dipartmento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Diabetologia ed Andrologia, University of Naples "Federico II", Naples, Italy
| | - Mariacarolina Salerno
- Department of Translational Medicine, Section of Pediatrics, University of Naples "Federico II", Naples, Italy
| | - Carmen Rosano
- Department of Translational Medicine, Section of Pediatrics, University of Naples "Federico II", Naples, Italy
| | - Barbara Brunetti
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", Section of Pediatrics, University of Salerno, Baronissi, Italy
| | - Pietro Strisciuglio
- Department of Translational Medicine, Section of Pediatrics, University of Naples "Federico II", Naples, Italy
| | - Annamaria Colao
- Dipartmento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Diabetologia ed Andrologia, University of Naples "Federico II", Naples, Italy
| | - Giancarlo Parenti
- Department of Translational Medicine, Section of Pediatrics, University of Naples "Federico II", Naples, Italy
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Daniela Melis
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", Section of Pediatrics, University of Salerno, Baronissi, Italy
| | - Terry G J Derks
- Section of Metabolic Diseases, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
10
|
Chu R, Wang Y, Kong J, Pan T, Yang Y, He J. Lipid nanoparticles as the drug carrier for targeted therapy of hepatic disorders. J Mater Chem B 2024; 12:4759-4784. [PMID: 38682294 DOI: 10.1039/d3tb02766j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 05/01/2024]
Abstract
The liver, a complex and vital organ in the human body, is susceptible to various diseases, including metabolic disorders, acute hepatitis, cirrhosis, and hepatocellular carcinoma. In recent decades, these diseases have significantly contributed to global morbidity and mortality. Currently, liver transplantation remains the most effective treatment for hepatic disorders. Nucleic acid therapeutics offer a selective approach to disease treatment through diverse mechanisms, enabling the regulation of relevant genes and providing a novel therapeutic avenue for hepatic disorders. It is expected that nucleic acid drugs will emerge as the third generation of pharmaceuticals, succeeding small molecule drugs and antibody drugs. Lipid nanoparticles (LNPs) represent a crucial technology in the field of drug delivery and constitute a significant advancement in gene therapies. Nucleic acids encapsulated in LNPs are shielded from the degradation of enzymes and effectively delivered to cells, where they are released and regulate specific genes. This paper provides a comprehensive review of the structure, composition, and applications of LNPs in the treatment of hepatic disorders and offers insights into prospects and challenges in the future development of LNPs.
Collapse
Affiliation(s)
- Runxuan Chu
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China.
| | - Yi Wang
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tung, Hong Kong SAR, P. R. China.
| | - Jianglong Kong
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tung, Hong Kong SAR, P. R. China.
| | - Ting Pan
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China.
- Department of Pharmaceutics School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Yani Yang
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China.
| | - Jun He
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China.
| |
Collapse
|
11
|
Nie H, Hu X, Xiong H, Zeng L, Chen W, Su T. Change and pathological significance of glycogen content in oral squamous cell carcinoma and oral submucous fibrosis. Tissue Cell 2024; 87:102337. [PMID: 38430849 DOI: 10.1016/j.tice.2024.102337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/07/2023] [Revised: 02/18/2024] [Accepted: 02/22/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVE This study aimed to investigate the change and pathological significance of glycogen content in oral squamous cell carcinoma (OSCC) and oral submucous fibrosis (OSF). METHODS AND MATERIALS 13 normal oral mucosa (NOM), 12 OSF mucosa, and 35 pairs of OSCC tissues and their corresponding adjacent mucosa tissues (AT) were collected from Xiangya Hospital for PAS staining to detect glycogen. Transcriptome sequencing data from OSCC were used to compare glycogen metabolism gene expression differences. Kaplan-Meier method was conducted to estimate Recurrence-free survival (RFS). RESULTS Glycogen levels were lower in OSF than in NOM and lower in OSCC than in AT. Transcriptome sequencing data analysis showed the expression of most glycogenolysis genes was increased and the expression of glycogen synthesis genes including PPP1R3C and GBE1 was decreased in OSCC tissues. High glycogen level was correlated with poor prognosis in OSCC patients under the background of OSF. CONCLUSION Glycogen may be used as a potential diagnostic biomolecule for OSF and OSCC, as well as a potential prognostic factor for OSCC in the context of OSF.
Collapse
Affiliation(s)
- Huanquan Nie
- Department of Oral and Maxillofacial Surgery, Centre of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Oral Cancer and Precancerous Lesions, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Xin Hu
- Department of Oral and Maxillofacial Surgery, Centre of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Oral Cancer and Precancerous Lesions, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Haofeng Xiong
- Department of Oral and Maxillofacial Surgery, Centre of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Oral Cancer and Precancerous Lesions, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Liujun Zeng
- Department of Oral and Maxillofacial Surgery, Centre of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Oral Cancer and Precancerous Lesions, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Wenxin Chen
- Department of Oral and Maxillofacial Surgery, Centre of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Oral Cancer and Precancerous Lesions, Central South University, China; Department of Orthodontics, Shanghai Huangpu District Dental Disease Prevention and Treatment Institute, Shanghai, China.
| | - Tong Su
- Department of Oral and Maxillofacial Surgery, Centre of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Oral Cancer and Precancerous Lesions, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China.
| |
Collapse
|
12
|
Zhang C, Huang W, Niu W, Yang H, Zheng Y, Gao X, Qiu X. Five genes identified as prognostic markers for colorectal cancer through the integration of genome-wide association study and expression quantitative trait loci data. Per Med 2024; 21:103-116. [PMID: 38380524 DOI: 10.2217/pme-2023-0103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/14/2023] [Accepted: 01/30/2024] [Indexed: 02/22/2024]
Abstract
Background: Colorectal cancer (CRC) is a prominent form of cancer globally, ranking second in terms of prevalence and serving as a leading cause of cancer-related deaths, but the underlying biological interpretation remains largely unknown. Methods: We used the summary data-based Mendelian randomization method to integrate CRC genome-wide association studies (ncase = 7062; ncontrol = 195,745) and expression quantitative trait loci summary data in peripheral whole blood (Consortium for Architecture of Gene Expression: n = 2765; Genotype-Tissue Expression [v8]: n = 755) and colon tissue (colon-transverse: n = 406; colon-sigmoid: n = 373) and identified related genes. Results: Genes ABTB1, CYP21A2, NLRP1, PHKG1 and PIP5K1C have emerged as significant prognostic markers for CRC patient survival. Functional analysis revealed their involvement in cancer cell migration and invasion mechanisms, providing valuable insights for the development of future anti-CRC drugs. Conclusion: We successfully identified five CRC risk genes, providing new insights and research directions for the effective mechanisms of CRC.
Collapse
Affiliation(s)
- Cuizhen Zhang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Wenjie Huang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Wanjie Niu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Huiying Yang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yingyi Zheng
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xuan Gao
- Outpatient and Emergency Management Office, National Children's Medical Center, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Xiaoyan Qiu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, 200040, China
| |
Collapse
|
13
|
Eldredge JA, Hardikar W. Current status and future directions of liver transplantation for metabolic liver disease in children. Pediatr Transplant 2024; 28:e14625. [PMID: 37859572 DOI: 10.1111/petr.14625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 05/31/2023] [Revised: 10/03/2023] [Accepted: 10/08/2023] [Indexed: 10/21/2023]
Abstract
Orthotopic liver transplantation (OLT) in the care of children with inborn errors of metabolism (IEM) is well established and represent the second most common indication for pediatric liver transplantation in most centers worldwide, behind biliary atresia. OLT offers cure of disease when a metabolic defect is confined to the liver, but may still be transformative on a patient's quality of life reducing the chance of metabolic crises causing neurological damage in children be with extrahepatic involvement and no "functional cure." Outcomes post-OLT for inborn errors of metabolism are generally excellent. However, this benefit must be balanced with consideration of a composite risk of morbidity, and commitment to a lifetime of post-transplant chronic disease management. An increasing number of transplant referrals for children with IEM has contributed to strain on graft access in many parts of the world. Pragmatic evaluation of IEM referrals is essential, particularly pertinent in cases where progression of extra-hepatic disease is anticipated, with long-term outcome expected to be poor. Decision to proceed with liver transplantation is highly individualized based on the child's dynamic risk-benefit profile, their family unit, and their treating multidisciplinary team. Also to be considered is the chance of future treatments, such as gene therapies, emerging in the medium term.
Collapse
Affiliation(s)
- Jessica A Eldredge
- Department of Gastroenterology, Hepatology and Clinical Nutrition, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Winita Hardikar
- Department of Gastroenterology, Hepatology and Clinical Nutrition, Royal Children's Hospital University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
14
|
Zamanfar D, Hashemi-Soteh SM, Ghazaiean M, Keyhanian E. Report of an Iranian child with chronic abdominal pain and constipation diagnosed as glycogen storage disease type IX: a case report. J Med Case Rep 2024; 18:14. [PMID: 38212860 PMCID: PMC10785502 DOI: 10.1186/s13256-023-04295-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/05/2022] [Accepted: 11/29/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND Glycogen storage disease type IX is a rare disorder that can cause a wide variety of symptoms depending on the specific deficiency of the phosphorylase kinase enzyme and the organs it affects. CASE PRESENTATION A 4-and-a-half-year-old Caucasian girl was referred to our clinic with a liver biopsy report indicating a diagnosis of glycogen storage disease. Prior to being referred to our clinic, the patient had been under the care of pediatric gastroenterologists. The patient's initial symptoms included chronic abdominal pain, constipation, and elevated liver transaminase. With the help of the pediatric gastroenterologists, cholestasis, Wilson disease, and autoimmune hepatitis were ruled out. Given that glycogen storage diseases type I and type III are the most common, we initially managed the patient with frequent feedings and a diet that included complex carbohydrates such as a corn starch supplement and a lactose restriction. Following an unfavorable growth velocity and hepatomegaly during the follow-up period, genetic analysis was conducted, which revealed a novel mutation of the phosphorylase kinase regulatory subunit beta gene- a c.C412T (P.Q138x) mutation. As the diagnosis of glycogen storage disease type IX was confirmed, the treatment regimen was altered to a high protein diet (more than 2 g/kg/day) and a low fat diet. CONCLUSION Given the mild and varied clinical manifestations of glycogen storage disease type IX, it is possible for the diagnosis to be overlooked. It is important to consider glycogen storage disease type IX in children who present with unexplained hepatomegaly and elevated transaminase levels. Furthermore, due to the distinct management of glycogen storage disease type IX compared with glycogen storage disease type I and glycogen storage disease type III, genetic analysis is essential for an accurate diagnosis.
Collapse
Affiliation(s)
- Daniel Zamanfar
- Department of Pediatric Endocrinology, Diabetes Reaserch Center of Mazandaran, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Seyed MohammadBagher Hashemi-Soteh
- Immunogenetic Research Center, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mobin Ghazaiean
- Gut and Liver Research Center, Non-Communicable Disease Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Elham Keyhanian
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran
| |
Collapse
|
15
|
Zhu H, Zhang T, Yuan H, Chen Y, Ding J, Ding H, Shi X, Gu D, Ma Y. A novel PHKA2 variant in a Chinese boy with glycogen storage diseases type IXa. Front Endocrinol (Lausanne) 2023; 14:1332450. [PMID: 38192425 PMCID: PMC10773889 DOI: 10.3389/fendo.2023.1332450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 11/03/2023] [Accepted: 12/04/2023] [Indexed: 01/10/2024] Open
Abstract
Background Glycogen storage diseases (GSDs) are a group of heterogeneous inherited metabolic disorders with an incidence of 4%-5%. There are 19 types of GSDs, making diagnosis one of the greatest challenges. Methods The proband and his parents were referred to our hospital for genetic diagnosis. Ultrasound screening suggested hepatomegaly. A novel insertion variant NM_000292 c.1155_1156insT (p. 386N>*) in PHKA2 gene was identified using trio whole exome sequencing (Trio-WES), which resulted in the codon of amino acid 386 from asparagine to termination (p. 386N>*). The 3D mutant protein structure was predicted using AlphaFold, and the results showed that the truncated PHKA2 protein contained 385 of the 1,235 amino acids of the mature protein. Conclusion We describe a previously unreported case of a GSDs IXa type Chinese boy caused by a novel PHKA2 variant. This clinical case contributes to the understanding of the characteristics of GSDs type IXa and expands the variants spectrum of genes related to GSDs type IXa. Our findings demonstrated the significance of genetic testing in the diagnosis of GSDs.
Collapse
Affiliation(s)
- Hongdan Zhu
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, Zhejiang, China
- Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - Tao Zhang
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, Zhejiang, China
- Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - Hua Yuan
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, Zhejiang, China
- Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - Yan Chen
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, Zhejiang, China
- Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - Jinlong Ding
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, Zhejiang, China
- Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - Haigang Ding
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, Zhejiang, China
- Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - Xiaoliang Shi
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, Zhejiang, China
- Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - Dalei Gu
- Beilun District People’s Hospital, Ningbo, Zhejiang, China
| | - Yingying Ma
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, Zhejiang, China
- Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| |
Collapse
|
16
|
Tummolo A, Carella R, De Giovanni D, Paterno G, Simonetti S, Tolomeo M, Leone P, Barile M. Micronutrient Deficiency in Inherited Metabolic Disorders Requiring Diet Regimen: A Brief Critical Review. Int J Mol Sci 2023; 24:17024. [PMID: 38069347 PMCID: PMC10707160 DOI: 10.3390/ijms242317024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/23/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Many inherited metabolic disorders (IMDs), including disorders of amino acid, fatty acid, and carbohydrate metabolism, are treated with a dietary reduction or exclusion of certain macronutrients, putting one at risk of a reduced intake of micronutrients. In this review, we aim to provide available evidence on the most common micronutrient deficits related to specific dietary approaches and on the management of their deficiency, in the meanwhile discussing the main critical points of each nutritional supplementation. The emerging concepts are that a great heterogeneity in clinical practice exists, as well as no univocal evidence on the most common micronutrient abnormalities. In phenylketonuria, for example, micronutrients are recommended to be supplemented through protein substitutes; however, not all formulas are equally supplemented and some of them are not added with micronutrients. Data on pyridoxine and riboflavin status in these patients are particularly scarce. In long-chain fatty acid oxidation disorders, no specific recommendations on micronutrient supplementation are available. Regarding carbohydrate metabolism disorders, the difficult-to-ascertain sugar content in supplementation formulas is still a matter of concern. A ketogenic diet may predispose one to both oligoelement deficits and their overload, and therefore deserves specific formulations. In conclusion, our overview points out the lack of unanimous approaches to micronutrient deficiencies, the need for specific formulations for IMDs, and the necessity of high-quality studies, particularly for some under-investigated deficits.
Collapse
Affiliation(s)
- Albina Tummolo
- Department of Metabolic Diseases, Clinical Genetics and Diabetology, Giovanni XXIII Children Hospital, Azienda Ospedaliero-Universitaria Consorziale, 70126 Bari, Italy; (R.C.); (D.D.G.); (G.P.)
| | - Rosa Carella
- Department of Metabolic Diseases, Clinical Genetics and Diabetology, Giovanni XXIII Children Hospital, Azienda Ospedaliero-Universitaria Consorziale, 70126 Bari, Italy; (R.C.); (D.D.G.); (G.P.)
| | - Donatella De Giovanni
- Department of Metabolic Diseases, Clinical Genetics and Diabetology, Giovanni XXIII Children Hospital, Azienda Ospedaliero-Universitaria Consorziale, 70126 Bari, Italy; (R.C.); (D.D.G.); (G.P.)
| | - Giulia Paterno
- Department of Metabolic Diseases, Clinical Genetics and Diabetology, Giovanni XXIII Children Hospital, Azienda Ospedaliero-Universitaria Consorziale, 70126 Bari, Italy; (R.C.); (D.D.G.); (G.P.)
| | - Simonetta Simonetti
- Regional Centre for Neonatal Screening, Department of Clinical Pathology and Neonatal Screening, Children’s Hospital “Giovanni XXIII”, Azienda Ospedaliero-Universitaria Consorziale, 70126 Bari, Italy;
| | - Maria Tolomeo
- Department of Biosciences, Biotechnology and Environment, University of Bari “A. Moro”, via Orabona 4, 70125 Bari, Italy; (M.T.); (P.L.)
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, via P. Bucci 4C, 87036 Arcavacata di Rende, Italy
| | - Piero Leone
- Department of Biosciences, Biotechnology and Environment, University of Bari “A. Moro”, via Orabona 4, 70125 Bari, Italy; (M.T.); (P.L.)
| | - Maria Barile
- Department of Biosciences, Biotechnology and Environment, University of Bari “A. Moro”, via Orabona 4, 70125 Bari, Italy; (M.T.); (P.L.)
| |
Collapse
|
17
|
Hyer E, Alexander C, Rand EB, Diamond T. Liver Biopsy Leads to Serendipitous Diagnosis of Glycogen Storage Disease Type IX in a Patient With Fontan-Associated Liver Disease. JPGN REPORTS 2023; 4:e377. [PMID: 38034453 PMCID: PMC10684236 DOI: 10.1097/pg9.0000000000000377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 03/02/2023] [Accepted: 08/16/2023] [Indexed: 12/02/2023]
Abstract
Fontan-associated liver disease (FALD) is a form of congestive hepatopathy resulting from Fontan palliation procedures in patients with single ventricle physiology. Although there is variation between pediatric centers, the surveillance for FALD may include liver biopsies for assessment of degree of fibrosis. Our report describes a 7-year-old girl with hypoplastic left heart syndrome who underwent Fontan palliation at age 2, and presented with disproportionate hepatomegaly, elevated liver enzymes, and increased stiffness on liver elastography. Liver biopsy showed diffuse hepatocellular cytoplasmic glycogenation, leading to the diagnosis of glycogen storage disease IX. This case demonstrates the importance of investigating unexpected physical exam findings and the potential for serendipitous benefit of liver biopsy in FALD.
Collapse
Affiliation(s)
- Erin Hyer
- From the Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA
- Drexel University College of Medicine, Philadelphia, PA
| | - Caitlin Alexander
- Department of Anatomic Pathology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Elizabeth B. Rand
- From the Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Tamir Diamond
- From the Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
18
|
Abstract
Glycogen storage diseases (GSDs) are a group of rare, monogenic disorders that share a defect in the synthesis or breakdown of glycogen. This Primer describes the multi-organ clinical features of hepatic GSDs and muscle GSDs, in addition to their epidemiology, biochemistry and mechanisms of disease, diagnosis, management, quality of life and future research directions. Some GSDs have available guidelines for diagnosis and management. Diagnostic considerations include phenotypic characterization, biomarkers, imaging, genetic testing, enzyme activity analysis and histology. Management includes surveillance for development of characteristic disease sequelae, avoidance of fasting in several hepatic GSDs, medically prescribed diets, appropriate exercise regimens and emergency letters. Specific therapeutic interventions are available for some diseases, such as enzyme replacement therapy to correct enzyme deficiency in Pompe disease and SGLT2 inhibitors for neutropenia and neutrophil dysfunction in GSD Ib. Progress in diagnosis, management and definitive therapies affects the natural course and hence morbidity and mortality. The natural history of GSDs is still being described. The quality of life of patients with these conditions varies, and standard sets of patient-centred outcomes have not yet been developed. The landscape of novel therapeutics and GSD clinical trials is vast, and emerging research is discussed herein.
Collapse
Affiliation(s)
- William B Hannah
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA.
| | - Terry G J Derks
- Division of Metabolic Diseases, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mitchell L Drumm
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Sarah C Grünert
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Medical Centre-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Priya S Kishnani
- Division of Medical Genetics, Department of Paediatrics, Duke University Medical Center, Durham, NC, USA
| | - John Vissing
- Copenhagen Neuromuscular Center, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
19
|
Quarta A, Iannucci D, Guarino M, Blasetti A, Chiarelli F. Hypoglycemia in Children: Major Endocrine-Metabolic Causes and Novel Therapeutic Perspectives. Nutrients 2023; 15:3544. [PMID: 37630734 PMCID: PMC10459037 DOI: 10.3390/nu15163544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/24/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Hypoglycemia is due to defects in the metabolic systems involved in the transition from the fed to the fasting state or in the hormone control of these systems. In children, hypoglycemia is considered a metabolic-endocrine emergency, because it may lead to brain injury, permanent neurological sequelae and, in rare cases, death. Symptoms are nonspecific, particularly in infants and young children. Diagnosis is based on laboratory investigations during a hypoglycemic event, but it may also require biochemical tests between episodes, dynamic endocrine tests and molecular genetics. This narrative review presents the age-related definitions of hypoglycemia, its pathophysiology and main causes, and discusses the current diagnostic and modern therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | | | - Francesco Chiarelli
- Department of Pediatrics, University of Chieti—Pescara, Gabriele D’Annunzio, 66100 Chieti, Italy; (A.Q.); (D.I.); (M.G.); (A.B.)
| |
Collapse
|
20
|
Abstract
Glycogen storage diseases (GSDs), also referred to as glycogenoses, are inherited metabolic disorders of glycogen metabolism caused by deficiency of enzymes or transporters involved in the synthesis or degradation of glycogen leading to aberrant storage and/or utilization. The overall estimated GSD incidence is 1 case per 20000-43000 live births. There are over 20 types of GSD including the subtypes. This heterogeneous group of rare diseases represents inborn errors of carbohydrate metabolism and are classified based on the deficient enzyme and affected tissues. GSDs primarily affect liver or muscle or both as glycogen is particularly abundant in these tissues. However, besides liver and skeletal muscle, depending on the affected enzyme and its expression in various tissues, multiorgan involvement including heart, kidney and/or brain may be seen. Although GSDs share similar clinical features to some extent, there is a wide spectrum of clinical phenotypes. Currently, the goal of treatment is to maintain glucose homeostasis by dietary management and the use of uncooked cornstarch. In addition to nutritional interventions, pharmacological treatment, physical and supportive therapies, enzyme replacement therapy (ERT) and organ transplantation are other treatment approaches for both disease manifestations and long-term complications. The lack of a specific therapy for GSDs has prompted efforts to develop new treatment strategies like gene therapy. Since early diagnosis and aggressive treatment are related to better prognosis, physicians should be aware of these conditions and include GSDs in the differential diagnosis of patients with relevant manifestations including fasting hypoglycemia, hepatomegaly, hypertransaminasemia, hyperlipidemia, exercise intolerance, muscle cramps/pain, rhabdomyolysis, and muscle weakness. Here, we aim to provide a comprehensive review of GSDs. This review provides general characteristics of all types of GSDs with a focus on those with liver involvement.
Collapse
Affiliation(s)
- Ersin Gümüş
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, Hacettepe University Faculty of Medicine, Ihsan Dogramaci Children’s Hospital, Ankara 06230, Turkey
| | - Hasan Özen
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, Hacettepe University Faculty of Medicine, Ihsan Dogramaci Children’s Hospital, Ankara 06230, Turkey
| |
Collapse
|
21
|
Lin WD, Tsai FJ, Wang CH. Identification of a novel deletion mutation in PHKA2 in a taiwanese patient with type IXa glycogen storage disease. Pediatr Neonatol 2023; 64:465-467. [PMID: 37280121 DOI: 10.1016/j.pedneo.2023.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 11/30/2022] [Revised: 03/04/2023] [Accepted: 03/28/2023] [Indexed: 06/08/2023] Open
Affiliation(s)
- Wei-De Lin
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan; School of Post Baccalaureate Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Fuu-Jen Tsai
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan; Division of Genetics and Metabolism, China Medical University Children's Hospital, Taichung, Taiwan; Department of Medical Genetics, China Medical University Hospital, Taichung, Taiwan; School of Chinese Medicine, China Medical University, Taichung, Taiwan; Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
| | - Chung-Hsing Wang
- Division of Genetics and Metabolism, China Medical University Children's Hospital, Taichung, Taiwan; School of Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
22
|
Hahn JW, Lee H, Seong MW, Kang GH, Moon JS, Ko JS. Clinical and genetic spectrum of GSD type 6 in Korea. Orphanet J Rare Dis 2023; 18:132. [PMID: 37264426 DOI: 10.1186/s13023-023-02750-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/20/2023] [Accepted: 05/18/2023] [Indexed: 06/03/2023] Open
Abstract
BACKGROUND Glycogen storage disease type VI (GSD VI) is a rare disease in which liver glycogen metabolism is impaired by mutations in the glycogen phosphorylase L (PYGL). This study aimed to examine the clinical features, genetic analyses, and long-term outcomes of patients with GSD VI in Korea. METHODS From January 2002 to November 2022, we retrospectively reviewed patients diagnosed with GSD VI using a gene panel at Seoul National University Hospital. We investigated the clinical profile, liver histology, molecular diagnosis, and long-term outcomes of patients with GSD VI. RESULTS Five patients were included in the study. The age at onset was 18-30 months (median, 21 months), and current age was 3.7-17 years (median, 11 years). All patients showed hepatomegaly, elevated liver transaminase activity, and hypertriglyceridaemia. Hypercholesterolaemia and fasting hypoglycaemia occurred in 60% and 40% of patients, respectively. Ten variants of PYGL were identified, of which six were novel: five missense (p.[Gly607Val], p.[Leu445Pro], p.[Gly695Glu], p.[Val828Gly], p.[Tyr158His]), and one frameshift (p.[Arg67AlafsTer34]). All patients were treated with a high-protein diet, and four also received corn starch. All patients showed improved liver function tests, hypertriglyceridaemia, hepatomegaly, and height z score. CONCLUSIONS The GSD gene panel is a useful diagnostic tool for confirming the presence of GSD VI. Genetic heterogeneity was observed in all patients with GSD VI. Increased liver enzyme levels, hypertriglyceridaemia, and height z score in patients with GSD VI improved during long-term follow-up.
Collapse
Affiliation(s)
- Jong Woo Hahn
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
- Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam, Gyeonggi- do, Korea
| | - Heerah Lee
- Department of Laboratory, Seoul National University College of Medicine, Seoul, Korea
| | - Moon Woo Seong
- Department of Laboratory, Seoul National University College of Medicine, Seoul, Korea
| | - Gyeong Hoon Kang
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Jin Soo Moon
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | - Jae Sung Ko
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
23
|
Tokatly Latzer I, Pearl PL. Treatment of neurometabolic epilepsies: Overview and recent advances. Epilepsy Behav 2023; 142:109181. [PMID: 37001467 DOI: 10.1016/j.yebeh.2023.109181] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 08/15/2022] [Revised: 03/11/2023] [Accepted: 03/12/2023] [Indexed: 05/08/2023]
Abstract
The rarity and heterogeneity of neurometabolic diseases make it challenging to reach evidence-based principles for their specific treatments. Indeed, current treatments for many of these diseases remain symptomatic and supportive. However, an ongoing scientific and medical revolution has led to dramatic breakthroughs in molecular sciences and genetics, revealing precise pathophysiologic mechanisms. Accordingly, this has led to significant progress in the development of novel therapeutic approaches aimed at treating epilepsy resulting from these conditions, as well as their other manifestations. We overview recent notable treatment advancements, from vitamins, trace minerals, and diets to unique medications targeting the elemental pathophysiology at a molecular or cellular level, including enzyme replacement therapy, enzyme enhancing therapy, antisense oligonucleotide therapy, stem cell transplantation, and gene therapy.
Collapse
Affiliation(s)
- Itay Tokatly Latzer
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Phillip L Pearl
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
24
|
Koch RL, Soler-Alfonso C, Kiely BT, Asai A, Smith AL, Bali DS, Kang PB, Landstrom AP, Akman HO, Burrow TA, Orthmann-Murphy JL, Goldman DS, Pendyal S, El-Gharbawy AH, Austin SL, Case LE, Schiffmann R, Hirano M, Kishnani PS. Diagnosis and management of glycogen storage disease type IV, including adult polyglucosan body disease: A clinical practice resource. Mol Genet Metab 2023; 138:107525. [PMID: 36796138 DOI: 10.1016/j.ymgme.2023.107525] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 12/12/2022] [Revised: 01/20/2023] [Accepted: 01/22/2023] [Indexed: 01/26/2023]
Abstract
Glycogen storage disease type IV (GSD IV) is an ultra-rare autosomal recessive disorder caused by pathogenic variants in GBE1 which results in reduced or deficient glycogen branching enzyme activity. Consequently, glycogen synthesis is impaired and leads to accumulation of poorly branched glycogen known as polyglucosan. GSD IV is characterized by a remarkable degree of phenotypic heterogeneity with presentations in utero, during infancy, early childhood, adolescence, or middle to late adulthood. The clinical continuum encompasses hepatic, cardiac, muscular, and neurologic manifestations that range in severity. The adult-onset form of GSD IV, referred to as adult polyglucosan body disease (APBD), is a neurodegenerative disease characterized by neurogenic bladder, spastic paraparesis, and peripheral neuropathy. There are currently no consensus guidelines for the diagnosis and management of these patients, resulting in high rates of misdiagnosis, delayed diagnosis, and lack of standardized clinical care. To address this, a group of experts from the United States developed a set of recommendations for the diagnosis and management of all clinical phenotypes of GSD IV, including APBD, to support clinicians and caregivers who provide long-term care for individuals with GSD IV. The educational resource includes practical steps to confirm a GSD IV diagnosis and best practices for medical management, including (a) imaging of the liver, heart, skeletal muscle, brain, and spine, (b) functional and neuromusculoskeletal assessments, (c) laboratory investigations, (d) liver and heart transplantation, and (e) long-term follow-up care. Remaining knowledge gaps are detailed to emphasize areas for improvement and future research.
Collapse
Affiliation(s)
- Rebecca L Koch
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA.
| | - Claudia Soler-Alfonso
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Bridget T Kiely
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Akihiro Asai
- Department of Pediatrics, University of Cincinnati Medical Center, Cincinnati, OH, USA; Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ariana L Smith
- Division of Urology, Department of Surgery, University of Pennsylvania Health System, Philadelphia, PA, USA
| | - Deeksha S Bali
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Peter B Kang
- Paul and Sheila Wellstone Muscular Dystrophy Center, Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Andrew P Landstrom
- Division of Cardiology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA; Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - H Orhan Akman
- Department of Neurology, Columbia University Irving Medical Center, New York City, NY, USA
| | - T Andrew Burrow
- Section of Genetics and Metabolism, Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock, AR, USA
| | | | - Deberah S Goldman
- Adult Polyglucosan Body Disease Research Foundation, Brooklyn, NY, USA
| | - Surekha Pendyal
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Areeg H El-Gharbawy
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Stephanie L Austin
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Laura E Case
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA; Doctor of Physical Therapy Division, Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | | | - Michio Hirano
- Department of Neurology, Columbia University Irving Medical Center, New York City, NY, USA
| | - Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
25
|
Colonetti K, Pinto E Vairo F, Siebert M, Nalin T, Poloni S, Fernando Wurdig Roesch L, Fischinger Moura de Souza C, Cabral Pinheiro F, Vanessa Doederlein Schwartz I. Cytokine profiling in patients with hepatic glycogen storage disease: Are there clues for unsolved aspects? Cytokine 2023; 162:156088. [PMID: 36462220 DOI: 10.1016/j.cyto.2022.156088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/05/2022] [Revised: 11/09/2022] [Accepted: 11/12/2022] [Indexed: 12/05/2022]
Abstract
INTRODUCTION Hepatic Glycogen Storage Diseases (GSD) are rare genetic disorders in which the gluconeogenesis pathway is impaired. Cytokines control virtually every aspect of physiology and may help to elucidate some unsolved questions about phenotypes presented by GSD patients. METHODS This was an exploratory study in which 27 GSD patients on treatment (Ia = 16, Ib = 06, III = 02, IXα = 03) and 24 healthy age- and sex-matched subjects had plasma samples tested for a panel of 20 cytokines (G-CSF,GM-CSF, IL-1α,IL-1β, IL-4, IL-6, IL-8, IL-10, IL-13, IL-17A, GRO, IP-10/CXCL10, MCP-1/CCL2, MIP-1α/CCL3, MIP-1β/CCL4, MDC/CCL22, IFN-γ, TNF-α, TNF-β, VEGF) through a multiplex kit and analyzed in comparison to controls and among patients, regarding to clinical features as anemia, hepatic adenocarcinoma and triglyceride levels. RESULTS Patients (GSD-Ia/III/IX) presented reduced levels of IL-4 (p = 0.040), MIP-1α/CCL3 (p = 0.003), MDC/CCL22 (p < 0.001), TNF-β (p = 0.045) and VEGF (p = 0.043) compared to controls. When different types of GSD were compared, G-CSF was higher in GSD-Ib than -Ia (p < 0.001) and than -III/IX (p = 0.033) patients; IL-10 was higher in GSD-Ib than in GSD-Ia patients (p = 0.019); and GSD-III/IX patients had increased levels of IP-10/CXCL10 than GSD-Ib patients (p = 0.019). When GSD-I patients were gathered into the same group and compared with GSD-III/IX patients, IP10/CXCL10 and MCP-1 were higher in the latter group (p = 0.005 and p = 0.013, respectively). GSD-I patients with anemia presented higher levels of IL-4 and MIP-1α in comparison with patients who had not. Triglyceride level was correlated with neutrophil count and MDC levels on GSD-Ia patients without HCA. CONCLUSION Altogether, altered levels of cytokines in GSD-I patients reflect an imbalance in immunoregulation process. This study also indicates that neutrophils and some cytokines are affected by triglyceride levels, and future studies on the theme should consider this variable.
Collapse
Affiliation(s)
- Karina Colonetti
- Post-Graduation Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratory of Basic Research and Advanced Investigations in Neurosciences (BRAIN), Hospital de Clínicas de Porto Alegre, PortoAlegre, RS, Brazil
| | - Filippo Pinto E Vairo
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA; Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| | - Marina Siebert
- Laboratory of Basic Research and Advanced Investigations in Neurosciences (BRAIN), Hospital de Clínicas de Porto Alegre, PortoAlegre, RS, Brazil; Post-Graduation Program in Sciences of Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratorial Research Unit, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Tatiéle Nalin
- Ultragenyx Brasil Farmacêutica Ltda, São Paulo, SP, Brazil
| | - Soraia Poloni
- Laboratory of Basic Research and Advanced Investigations in Neurosciences (BRAIN), Hospital de Clínicas de Porto Alegre, PortoAlegre, RS, Brazil
| | - Luiz Fernando Wurdig Roesch
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, USA
| | - Carolina Fischinger Moura de Souza
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-Graduation Program in Child and Adolescent Health, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Franciele Cabral Pinheiro
- Post-Graduation Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratory of Basic Research and Advanced Investigations in Neurosciences (BRAIN), Hospital de Clínicas de Porto Alegre, PortoAlegre, RS, Brazil; Universidade Federal do Pampa, Itaqui, RS, Brazil
| | - Ida Vanessa Doederlein Schwartz
- Post-Graduation Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratory of Basic Research and Advanced Investigations in Neurosciences (BRAIN), Hospital de Clínicas de Porto Alegre, PortoAlegre, RS, Brazil; Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
| |
Collapse
|
26
|
Integrating Network Pharmacology and Transcriptomic Strategies to Explore the Pharmacological Mechanism of Hydroxysafflor Yellow A in Delaying Liver Aging. Int J Mol Sci 2022; 23:ijms232214281. [PMID: 36430769 PMCID: PMC9697017 DOI: 10.3390/ijms232214281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/16/2022] [Revised: 11/03/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022] Open
Abstract
Aging affects the structure and function of the liver. Hydroxysafflor yellow A (HSYA) effectively improves liver aging (LA) in mice, but the potential mechanisms require further exploration. In this study, an integrated approach combining network pharmacology and transcriptomics was used to elucidate the potential mechanisms of HSYA delay of LA. The targets of HSYA were predicted using the PharmMapper, SwissTargetPrediction, and CTD databases, and the targets of LA were collected from the GeneCards database. An ontology (GO) analysis and a Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway annotation of genes related to HSYA delay of LA were performed using the DAVID database, and Cytoscape software was used to construct an HSYA target pathway network. The BMKCloud platform was used to sequence mRNA from mouse liver tissue, screen differentially expressed genes (DEGs) that were altered by HSYA, and enrich their biological functions and signaling pathways through the OmicShare database. The results of the network pharmacology and transcriptomic analyses were combined. Then, quantitative real-time PCR (qRT-PCR) and Western blot experiments were used to further verify the prediction results. Finally, the interactions between HSYA and key targets were assessed by molecular docking. The results showed that 199 potentially targeted genes according to network pharmacology and 480 DEGs according to transcriptomics were involved in the effects of HSYA against LA. An integrated analysis revealed that four key targets, including HSP90AA1, ATP2A1, NOS1 and CRAT, as well as their three related pathways (the calcium signaling pathway, estrogen signaling pathway and cGMP-PKG signaling pathway), were closely related to the therapeutic effects of HSYA. A gene and protein expression analysis revealed that HSYA significantly inhibited the expressions of HSP90AA1, ATP2A1 and NOS1 in the liver tissue of aging mice. The molecular docking results showed that HSYA had high affinities with the HSP90AA1, ATP2A1 and NOS1 targets. Our data demonstrate that HSYA may delay LA in mice by inhibiting the expressions of HSP90AA1, ATP2A1 and NOS1 and regulating the calcium signaling pathway, the estrogen signaling pathway, and the cGMP-PKG signaling pathway.
Collapse
|
27
|
Venema A, Peeks F, Rossi A, Jager EA, Derks TGJ. Towards values-based healthcare for inherited metabolic disorders: An overview of current practices for persons with liver glycogen storage disease and fatty acid oxidation disorders. J Inherit Metab Dis 2022; 45:1018-1027. [PMID: 36088581 PMCID: PMC9828459 DOI: 10.1002/jimd.12555] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 02/20/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 01/12/2023]
Abstract
Value-based healthcare (VBHC) intends to achieve better outcomes for patients, to improve quality of patient care, with reduced costs. Four dimensions define a model of intimately related value-pillars: personal value, allocative value, technical value, and societal value. VBHC is mostly applied in common diseases, and there are fundamental challenges in applying VBHC strategies to low volume, high complex healthcare situations, such as rare diseases, including inherited metabolic disorders. This article summarizes current practices at various academical domains (i.e., research, healthcare, education, and training) that (aim to) increase values at various value-pillars for persons with liver glycogen storage diseases or fatty acid oxidation disorders and their families. Future perspectives may include facilitating virtual networks to function as integrated practice units, improving measurement of outcomes, and creating information technology platforms to overcome the ethical, legal, societal, and technical challenges of data sharing for healthcare and research purposes.
Collapse
Affiliation(s)
- Annieke Venema
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Centre GroningenUniversity of GroningenGroningenThe Netherlands
| | - Fabian Peeks
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Centre GroningenUniversity of GroningenGroningenThe Netherlands
| | - Alessandro Rossi
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Centre GroningenUniversity of GroningenGroningenThe Netherlands
- Department of Translational Medicine, Section of PediatricsUniversity of Naples “Federico II”NaplesItaly
| | - Emmalie A. Jager
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Centre GroningenUniversity of GroningenGroningenThe Netherlands
| | - Terry G. J. Derks
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Centre GroningenUniversity of GroningenGroningenThe Netherlands
| |
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW Glycogen storage disease is a group of disorders primarily characterized by hepatomegaly and fasting hypoglycemia. This group of disorders may also affect the muscle, kidneys, and neurodevelopment. With an overall prevalence of 1 : 20 000, GSDs are disorders that clinicians should diagnose in a timely manner because adequate management can prevent complications, such as neurodevelopmental delay and liver disease [1] . As there are numerous types of GSDs, being able to distinguish one type from another can be overwhelming. In this review, we focus on hepatic GSDs to provide a concise review of clinical presentation, diagnosis, and current management. RECENT FINDINGS GSDs are considered rare disorders, and one of the main challenges is the delay in diagnosis, misdiagnosis, or under diagnosis. However, with molecular genetic testing now readily available, confirming the diagnosis is no longer as difficult or invasive as it was in the past. SUMMARY Current therapy for this group of disorders requires maintaining stable glucose levels. Avoiding hypoglycemia, as well as hyperglycemia, is critical in managing these patients. Being able to distinguish the types of GSDs and understanding the specific treatments for each enzymatic defect will optimize patient care.
Collapse
Affiliation(s)
- Tiffany L Freeney Wright
- Division of Pediatric Gastroenterology, Nutrition and Hepatology, Department of Pediatrics, University of Texas Southwestern Medical Center
- Children's Medical Center Dallas
| | - Luis A Umaña
- Children's Medical Center Dallas
- Division of Pediatric Genetics and Metabolism, Department of Pediatrics, University of Texas Southwestern Medical Center, Texas, USA
| | - Charina M Ramirez
- Division of Pediatric Gastroenterology, Nutrition and Hepatology, Department of Pediatrics, University of Texas Southwestern Medical Center
- Children's Medical Center Dallas
| |
Collapse
|
29
|
Arends CJ, Wilson LH, Estrella A, Kwon OS, Weinstein DA, Lee YM. A Mouse Model of Glycogen Storage Disease Type IX-Beta: A Role for Phkb in Glycogenolysis. Int J Mol Sci 2022; 23:ijms23179944. [PMID: 36077341 PMCID: PMC9456097 DOI: 10.3390/ijms23179944] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/10/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Glycogen storage disease type IX (GSD-IX) constitutes nearly a quarter of all GSDs. This ketotic form of GSD is caused by mutations in phosphorylase kinase (PhK), which is composed of four subunits (α, β, γ, δ). PhK is required for the activation of the liver isoform of glycogen phosphorylase (PYGL), which generates free glucose-1-phosphate monomers to be used as energy via cleavage of the α -(1,4) glycosidic linkages in glycogen chains. Mutations in any of the PhK subunits can negatively affect the regulatory and catalytic activity of PhK during glycogenolysis. To understand the pathogenesis of GSD-IX-beta, we characterized a newly created PHKB knockout (Phkb−/−) mouse model. In this study, we assessed fasting blood glucose and ketone levels, serum metabolite concentrations, glycogen phosphorylase activity, and gene expression of gluconeogenic genes and fibrotic genes. Phkb−/− mice displayed hepatomegaly with lower fasting blood glucose concentrations. Phkb−/− mice showed partial liver glycogen phosphorylase activity and increased sensitivity to pyruvate, indicative of partial glycogenolytic activity and upregulation of gluconeogenesis. Additionally, gene expression analysis demonstrated increased lipid metabolism in Phkb−/− mice. Gene expression analysis and liver histology in the livers of old Phkb−/− mice (>40 weeks) showed minimal profibrogenic features when analyzed with age-matched wild-type (WT) mice. Collectively, the Phkb−/− mouse recapitulates mild clinical features in patients with GSD-IX-beta. Metabolic and molecular analysis confirmed that Phkb−/− mice were capable of sustaining energy homeostasis during prolonged fasting by using partial glycogenolysis, increased gluconeogenesis, and potentially fatty acid oxidation in the liver.
Collapse
Affiliation(s)
- Charles J. Arends
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT 06030, USA
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Lane H. Wilson
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT 06030, USA
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ana Estrella
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Oh Sung Kwon
- Department of Kinesiology, University of Connecticut, Storrs, CT 06269, USA
- Department of Orthopaedic Surgery and Center on Aging, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - David A. Weinstein
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Young Mok Lee
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT 06030, USA
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
- Correspondence:
| |
Collapse
|
30
|
Barot S, Stephenson OJ, Priya Vemana H, Yadav A, Bhutkar S, Trombetta LD, Dukhande VV. Metabolic alterations and mitochondrial dysfunction underlie hepatocellular carcinoma cell death induced by a glycogen metabolic inhibitor. Biochem Pharmacol 2022; 203:115201. [PMID: 35926650 PMCID: PMC10039449 DOI: 10.1016/j.bcp.2022.115201] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/19/2022] [Revised: 07/19/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related deaths. There is an urgent need for new targets to treat HCC due to limited treatment options and drug resistance. Many cancer cells are known to have high amount of glycogen than their tissue of origin and inhibition of glycogen catabolism induces cancer cell death by apoptosis. To further understand the role of glycogen in HCC and target it for pharmacotherapy, we studied metabolic adaptations and mitochondrial function in HepG2 cells after pharmacological inhibition of glycogen phosphorylase (GP) by CP-91149 (CP). GP inhibition increased the glycogen levels in HepG2 cells without affecting overall glucose uptake. Glycolytic capacity and importantly glycolytic reserve decreased significantly. Electron microscopy revealed that CP treatment altered mitochondrial morphology leading to mitochondrial swelling with less defined cristae. A concomitant decrease in mitochondrial oxygen consumption and mitochondria-linked ATP generation was observed. Metabolomics and enzyme activity / expression studies showed a decrease in the pentose phosphate pathway. In addition, CP treatment decreased the growth of HepG2 3D tumor spheroids in a dose- and time-dependent manner. Taken together, our study provides insights into metabolic alterations and mitochondrial dysfunction accompanying apoptosis in HepG2 cells upon GP inhibition. Our study can aid in the understanding of the mechanism and development of metabolic inhibitors to treat HCC.
Collapse
Affiliation(s)
- Shrikant Barot
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Olivia J Stephenson
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Hari Priya Vemana
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Anjali Yadav
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Shraddha Bhutkar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Louis D Trombetta
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Vikas V Dukhande
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| |
Collapse
|
31
|
Dong R, Wei X, Zhang K, Song F, Lv Y, Gao M, Wang D, Ma J, Gai Z, Liu Y. Genotypic and phenotypic characteristics of 12 chinese children with glycogen storage diseases. Front Genet 2022; 13:932760. [PMID: 36105079 PMCID: PMC9465291 DOI: 10.3389/fgene.2022.932760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/05/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Glycogen storage diseases (GSDs) are known as a group of disorders characterized by genetic errors leading to accumulation of glycogen in various tissues. Since different types of GSD can sometimes be clinically indistinguishable, next generation sequencing is becoming a powerful tool for clinical diagnosis. Methods: 12 patients with suspected GSDs and their parents were enrolled in this study. The clinical and laboratory data of the patients were reviewed. Causative gene variants were identified in the patients using whole exome sequencing (WES) and verified by Sanger sequencing. Results: Genetic testing and analysis showed that 7 patients were diagnosed with GSD II (Pompe disease), 2 patients with GSD III, 1 patient with GSD VI, and 2 patients with GSD IXα. A total number of 18 variants were identified in 12 patients including 11 variants in GAA gene, 3 variants in AGL gene, 2 variants in PYGL gene and 2 variants in PHKA2 gene, of which 9 variants were reported and 9 variants were novel. SIFT, Polyphen-2, Mutation Taster, and REVEL predicted the novel variants (except GAA c.1052_1075 + 47del) to be disease-causing. The 3D structures of wild/mutant type GAA protein were predicted indicating that variants p. Trp621Gly, p. Pro541Leu, p. Ser800Ile and p. Gly293Trp might affect the proteins function via destroying hydrogen bonds or conformational constraints. Neither liver size nor laboratory findings allow for a differentiation among GSD III, GSD VI and GSD IXα. Conclusion: Our study expanded the variation spectrum of genes associated with GSDs. WES, in combination with clinical, biochemical, and pathological hallmarks, could provide accurate results for diagnosing and sub-typing GSD and related diseases in clinical setting.
Collapse
Affiliation(s)
- Rui Dong
- Pediatric Research Institute, Children’s Hospital Affiliated to Shandong University (Jinan Children’s Hospital), Jinan, China
- Shandong Provincial Clinical Research Center for Children’s Health and Disease, Jinan, China
| | - Xuxia Wei
- Shandong Provincial Clinical Research Center for Children’s Health and Disease, Jinan, China
- Gastroenterology, Children’s Hospital Affiliated to Shandong University (Jinan Children’s Hospital), Jinan, China
| | - Kaihui Zhang
- Pediatric Research Institute, Children’s Hospital Affiliated to Shandong University (Jinan Children’s Hospital), Jinan, China
- Shandong Provincial Clinical Research Center for Children’s Health and Disease, Jinan, China
| | - Fengling Song
- Shandong Provincial Clinical Research Center for Children’s Health and Disease, Jinan, China
- Children’s Health Department, Children’s Hospital Affiliated to Shandong University (Jinan Children’s Hospital), Jinan, China
| | - Yuqiang Lv
- Pediatric Research Institute, Children’s Hospital Affiliated to Shandong University (Jinan Children’s Hospital), Jinan, China
- Shandong Provincial Clinical Research Center for Children’s Health and Disease, Jinan, China
| | - Min Gao
- Pediatric Research Institute, Children’s Hospital Affiliated to Shandong University (Jinan Children’s Hospital), Jinan, China
- Shandong Provincial Clinical Research Center for Children’s Health and Disease, Jinan, China
| | - Dong Wang
- Pediatric Research Institute, Children’s Hospital Affiliated to Shandong University (Jinan Children’s Hospital), Jinan, China
- Shandong Provincial Clinical Research Center for Children’s Health and Disease, Jinan, China
| | - Jian Ma
- Pediatric Research Institute, Children’s Hospital Affiliated to Shandong University (Jinan Children’s Hospital), Jinan, China
- Shandong Provincial Clinical Research Center for Children’s Health and Disease, Jinan, China
| | - Zhongtao Gai
- Pediatric Research Institute, Children’s Hospital Affiliated to Shandong University (Jinan Children’s Hospital), Jinan, China
- Shandong Provincial Clinical Research Center for Children’s Health and Disease, Jinan, China
- *Correspondence: Zhongtao Gai, ; Yi Liu,
| | - Yi Liu
- Pediatric Research Institute, Children’s Hospital Affiliated to Shandong University (Jinan Children’s Hospital), Jinan, China
- Shandong Provincial Clinical Research Center for Children’s Health and Disease, Jinan, China
- *Correspondence: Zhongtao Gai, ; Yi Liu,
| |
Collapse
|
32
|
Huang K, Duan HQ, Li QX, Luo YB, Bi FF, Yang H. Expanding the clinicopathological-genetic spectrum of glycogen storage disease type IXd by a Chinese neuromuscular center. Front Neurol 2022; 13:945280. [PMID: 36034300 PMCID: PMC9406516 DOI: 10.3389/fneur.2022.945280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/16/2022] [Accepted: 07/19/2022] [Indexed: 11/29/2022] Open
Abstract
Background Glycogen storage disease (GSDs) is characterized by abnormally inherited glycogen metabolism. GSD IXd, which is caused by mutations in the PHKA1 gene, is an X-linked rare disease with mild myopathic symptoms. To date, only 13 patients with GSD IXd have been reported. In this study, we aimed to expand the clinicopathological-genetic spectrum of GSD IXd at a neuromuscular center in China. Methods Data on patients diagnosed with GSD IXd at our neuromuscular center were collected retrospectively. Clinical features, electrophysiology, muscle pathology, and genetic information were analyzed. Results Between 2015 and 2021, three patients were diagnosed with GSD IXd based on clinical manifestations, pathological findings, and genetic testing. One patient presented with mitochondrial myopathy. All patients exhibited muscle weakness and elevated levels of creatine kinase. Electromyography-detected myopathic changes were found in two patients, whereas one patient refused to undergo this examination. Pathological examinations in all patients revealed subsarcolemmal accumulation of glycogen under PAS staining. All patients had mutations in the PHKA1 gene and the patient with mitochondrial myopathy also had a mutation in the MT-TL1 gene. Conclusion Our study expands the clinicogenotype and phenotype of GSD IXd in a Chinese population. Our study also expands the known mutation spectrum for GSD IXd, contributing to a better characterization and understanding of this ultrarare neuromuscular disorder.
Collapse
Affiliation(s)
- Kun Huang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Hui-Qian Duan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Qiu-Xiang Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yue-Bei Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Fang-Fang Bi
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Huan Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- *Correspondence: Huan Yang
| |
Collapse
|
33
|
Tagliaferri F, Massese M, Russo L, Commone A, Gasperini S, Pretese R, Dionisi-Vici C, Maiorana A. Hepatic glycogen storage diseases type 0, VI and IX: description of an italian cohort. Orphanet J Rare Dis 2022; 17:285. [PMID: 35854365 PMCID: PMC9295101 DOI: 10.1186/s13023-022-02431-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/18/2022] [Accepted: 06/30/2022] [Indexed: 11/10/2022] Open
Abstract
Background Glycogen storage disease (GSD) type 0, VI and IX are inborn errors of metabolism involving hepatic glycogen synthesis and degradation. We performed a characterization of a large Italian cohort of 30 patients with GSD type 0a, VI, IXa, IXb and IXc. A retrospective evaluation of genetical, auxological and endocrinological data, biochemical tests, and nutritional intakes was assessed. Eventual findings of overweight/obesity and insulin-resistance were correlated with diet composition. Results Six GSD-0a, 1 GSD-VI, and 23 GSD-IX patients were enrolled, with an age of presentation from 0 to 72 months (median 14 months). Diagnosis was made at a median age of 30 months, with a median diagnostic delay of 11 months and a median follow-up of 66 months. From first to last visit, patients gained a median height of 0.6 SDS (from − 1.1 to 2.1 SDS) and a median weight of 0.5 SDS (from − 2.5 to 3.3 SDS); mean and minimal glucose values significant improved (p < 0.05). With respect to dietary intakes, protein intake (g/kg) and protein intake (g/kg)/RDA ratio directly correlated with the glucose/insulin ratio (p < 0.05) and inversely correlated with HOMA-IR (Homeostasis model assessment of insulin resistance, p < 0.05), BMI SDS (p < 0.05) and %ibw (ideal body weight percentage, p < 0.01). Conclusion A prompt establishment of specific nutritional therapy allowed to preserve growth, improve glycemic control and prevent liver complication, during childhood. Remarkably, the administration of a high protein diet appeared to have a protective effect against overweight/obesity and insulin-resistance.
Collapse
Affiliation(s)
- Francesco Tagliaferri
- Division of Metabolism, Department of Pediatric Subspecialties, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,SCDU of Pediatrics, Azienda Ospedaliero-Universitaria Maggiore Della Carità, University of Piemonte Orientale, Novara, Italy
| | - Miriam Massese
- Division of Metabolism, Department of Pediatric Subspecialties, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Luisa Russo
- Division of Metabolism, Department of Pediatric Subspecialties, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Anna Commone
- Division of Metabolism, Department of Pediatric Subspecialties, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Serena Gasperini
- Metabolic Unit Rare Disease, Pediatric Department, Fondazione MBBM, San Gerardo Hospital, Monza, Italy
| | - Roberta Pretese
- Metabolic Unit Rare Disease, Pediatric Department, Fondazione MBBM, San Gerardo Hospital, Monza, Italy
| | - Carlo Dionisi-Vici
- Division of Metabolism, Department of Pediatric Subspecialties, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Arianna Maiorana
- Division of Metabolism, Department of Pediatric Subspecialties, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| |
Collapse
|
34
|
Kumar TV, Bhat M, Narayanachar SG, Narayan V, Srikanth AK, Anikar S, Shetty S. Molecular and clinical profiling in a large cohort of Asian Indians with glycogen storage disorders. PLoS One 2022; 17:e0270373. [PMID: 35834487 PMCID: PMC9282608 DOI: 10.1371/journal.pone.0270373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/09/2022] [Accepted: 06/08/2022] [Indexed: 11/18/2022] Open
Abstract
Glycogen storage disorders occur due to enzyme deficiencies in the glycogenolysis and gluconeogenesis pathway, encoded by 26 genes. GSD’s present with overlapping phenotypes with variable severity. In this series, 57 individuals were molecularly confirmed for 7 GSD subtypes and their demographic data, clinical profiles and genotype-phenotype co-relations are studied. Genomic DNA from venous blood samples was isolated from clinically affected individuals. Targeted gene panel sequencing covering 23 genes and Sanger sequencing were employed. Various bioinformatic tools were used to predict pathogenicity for new variations. Close parental consanguinity was seen in 76%. Forty-nine pathogenic variations were detected of which 27 were novel. Variations were spread across GSDIa, Ib, III, VI, IXa, b and c. The largest subgroup was GSDIII in 28 individuals with 24 variations (12 novel) in AGL. The 1620+1G>C intronic variation was observed in 5 with GSDVI (PYGL). A total of eleven GSDIX are described with the first Indian report of type IXb. This is the largest study of GSDs from India. High levels of consanguinity in the local population and employment of targeted sequencing panels accounted for the range of GSDs reported here.
Collapse
Affiliation(s)
| | - Meenakshi Bhat
- Clinical Genetics, Centre for Human Genetics, Bengaluru, India
- Pediatric Genetics, Indira Gandhi Institute of Child Health, Bengaluru, India
| | | | - Vinu Narayan
- Clinical Genetics, Centre for Human Genetics, Bengaluru, India
| | | | - Swathi Anikar
- Molecular Genetics, Centre for Human Genetics, Bengaluru, India
| | - Swathi Shetty
- Molecular Genetics, Centre for Human Genetics, Bengaluru, India
- * E-mail:
| |
Collapse
|
35
|
Massese M, Tagliaferri F, Dionisi-Vici C, Maiorana A. Glycogen storage diseases with liver involvement: a literature review of GSD type 0, IV, VI, IX and XI. Orphanet J Rare Dis 2022; 17:241. [PMID: 35725468 PMCID: PMC9208159 DOI: 10.1186/s13023-022-02387-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/24/2022] [Accepted: 06/06/2022] [Indexed: 12/31/2022] Open
Abstract
Background Glycogen storage diseases (GSDs) with liver involvement are classified into types 0, I, III, IV, VI, IX and XI, depending on the affected enzyme. Hypoglycemia and hepatomegaly are hallmarks of disease, but muscular and renal tubular involvement, dyslipidemia and osteopenia can develop. Considering the paucity of literature available, herein we provide a narrative review of these latter forms of GSDs. Main body Diagnosis is based on clinical manifestations and laboratory test results, but molecular analysis is often necessary to distinguish the various forms, whose presentation can be similar. Compared to GSD type I and III, which are characterized by a more severe impact on metabolic and glycemic homeostasis, GSD type 0, VI, IX and XI are usually known to be responsive to the nutritional treatment for achieving a balanced metabolic homeostasis in the pediatric age. However, some patients can exhibit a more severe phenotype and an important progression of the liver and muscular disease. The effects of dietary adjustments in GSD type IV are encouraging, but data are limited. Conclusions Early diagnosis allows a good metabolic control, with improvement of quality of life and prognosis, therefore we underline the importance of building a proper knowledge among physicians about these rare conditions. Regular monitoring is necessary to restrain disease progression and complications.
Collapse
Affiliation(s)
- Miriam Massese
- Division of Metabolism, Department of Pediatric Subspecialties, Ospedale Pediatrico Bambino Gesù, IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy.,Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Francesco Tagliaferri
- Division of Metabolism, Department of Pediatric Subspecialties, Ospedale Pediatrico Bambino Gesù, IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy.,SCDU of Pediatrics, Azienda Ospedaliero-Universitaria Maggiore Della Carità, University of Piemonte Orientale, Novara, Italy
| | - Carlo Dionisi-Vici
- Division of Metabolism, Department of Pediatric Subspecialties, Ospedale Pediatrico Bambino Gesù, IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - Arianna Maiorana
- Division of Metabolism, Department of Pediatric Subspecialties, Ospedale Pediatrico Bambino Gesù, IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy.
| |
Collapse
|
36
|
dos Santos BB, Colonetti K, Nalin T, de Oliveira BM, de Souza CF, Spritzer PM, Schwartz IV. Body composition in patients with hepatic glycogen storage diseases. Nutrition 2022; 103-104:111763. [DOI: 10.1016/j.nut.2022.111763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/27/2021] [Revised: 05/14/2022] [Accepted: 05/31/2022] [Indexed: 10/31/2022]
|
37
|
Shao Y, Li T, Jiang M, Xu J, Huang Y, Li X, Zheng R, Liu L. A very rare case report of glycogen storage disease type IXc with novel PHKG2 variants. BMC Pediatr 2022; 22:267. [PMID: 35549678 PMCID: PMC9097106 DOI: 10.1186/s12887-021-03055-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 06/17/2021] [Accepted: 12/05/2021] [Indexed: 12/02/2022] Open
Abstract
Background Pathogenic mutations in the PHKG2 are associated with a very rare disease—glycogen storage disease IXc (GSD-IXc)—and are characterized by severe liver disease. Case presentation Here, we report a patient with jaundice, hypoglycaemia, growth retardation, progressive increase in liver transaminase and prominent hepatomegaly from the neonatal period. Genetic testing revealed two novel, previously unreported PHKG2 mutations (F233S and R320DfsX5). Functional experiments indicated that both F223S and R320DfsX5 lead to a decrease in key phosphorylase b kinase enzyme activity. With raw cornstarch therapy, hypoglycaemia and lactic acidosis were ameliorated and serum aminotransferases decreased. Conclusion These findings expand the gene spectrum and contribute to the interpretation of clinical presentations of these two novel PHKG2 mutations. Supplementary Information The online version contains supplementary material available at 10.1186/s12887-021-03055-7.
Collapse
Affiliation(s)
- Yongxian Shao
- Department of Pediatric Endocrinology and Genetic Metabolism, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Taolin Li
- Department of Pediatric Endocrinology and Genetic Metabolism, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Minyan Jiang
- Department of Pediatric Endocrinology and Genetic Metabolism, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jianan Xu
- Department of Pediatric Endocrinology and Genetic Metabolism, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yonglan Huang
- Department of Pediatric Endocrinology and Genetic Metabolism, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiuzhen Li
- Department of Pediatric Endocrinology and Genetic Metabolism, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Ruidan Zheng
- Department of Pediatric Endocrinology and Genetic Metabolism, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Li Liu
- Department of Pediatric Endocrinology and Genetic Metabolism, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
38
|
İnci A, Kılıç Yıldırım G, Cengiz Ergin FB, Sarı S, Eğritaş Gürkan Ö, Okur İ, Biberoğlu G, Bükülmez A, Ezgü FS, Dalgıç B, Tümer L. Expected or unexpected clinical findings in liver glycogen storage disease type IX: distinct clinical and molecular variability. J Pediatr Endocrinol Metab 2022; 35:451-462. [PMID: 35038814 DOI: 10.1515/jpem-2021-0278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 04/16/2021] [Accepted: 12/22/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES To reveal the different clinical presentations of liver glycogen storage disease type IX (GSD IX), which is a clinically and genetically heterogeneous type of glycogenosis. METHODS The data from the electronic hospital records of 25 patients diagnosed with liver GSD IX was reviewed. Symptoms, clinical findings, and laboratory and molecular analysis were assessed. RESULTS Of the patients, 10 had complaints of short stature in the initial presentation additionally other clinical findings. Elevated serum transaminases were found in 20 patients, and hepatomegaly was found in 22 patients. Interestingly, three patients were referred due to neurodevelopmental delay and hypotonia, while one was referred for only autism. One patient who presented with neurodevelopmental delay developed hepatomegaly and elevated transaminases during the disease later on. Three of the patients had low hemoglobin A1C and fructosamine values that were near the lowest reference range. Two patients had left ventricular hypertrophy. Three patients developed osteopenia during follow-up, and one patient had osteoporosis after puberty. The most common gene variant, PHKA2, was observed in 16 patients, 10 variants were novel and six variants were defined before. Six patients had variants in PHKG2, two variants were not defined before and four variants were defined before. PHKB variants were found in three patients. One patient had two novel splice site mutations in trans position. It was revealed that one novel homozygous variant and one defined homozygous variant were found in PHKB. CONCLUSIONS This study revealed that GSD IX may present with only hypotonia and neurodevelopmental delay without liver involvement in the early infantile period. It should be emphasized that although liver GSDIX is thought of as a benign disease, it might present with multisystemic involvement and patients should be screened with echocardiography, bone mineral densitometry, and psychometric evaluation.
Collapse
Affiliation(s)
- Aslı İnci
- Department of Pediatric Metabolism and Nutrition, Gazi University School of Medicine, Ankara, Turkey
| | - Gonca Kılıç Yıldırım
- Department of Pediatric Metabolism and Nutrition, Osmangazi University School of Medicine, Eskisehir, Turkey
| | - Filiz Başak Cengiz Ergin
- Department of Pediatric Metabolism and Nutrition, Gazi University School of Medicine, Ankara, Turkey
| | - Sinan Sarı
- Department of Pediatric Gastroenterology and Hepatology, Gazi University School of Medicine, Ankara, Turkey
| | - Ödül Eğritaş Gürkan
- Department of Pediatric Gastroenterology and Hepatology, Gazi University School of Medicine, Ankara, Turkey
| | - İlyas Okur
- Department of Pediatric Metabolism and Nutrition, Gazi University School of Medicine, Ankara, Turkey
| | - Gürsel Biberoğlu
- Department of Pediatric Metabolism and Nutrition, Gazi University School of Medicine, Ankara, Turkey
| | - Ayşegül Bükülmez
- Department of Pediatric Gastroenterology and Hepatology, Afyon Kocatepe University School of Medicine, Afyon, Turkey
| | - Fatih Süheyl Ezgü
- Department of Pediatric Metabolism and Nutrition, Gazi University School of Medicine, Ankara, Turkey
| | - Buket Dalgıç
- Department of Pediatric Gastroenterology and Hepatology, Gazi University School of Medicine, Ankara, Turkey
| | - Leyla Tümer
- Department of Pediatric Metabolism and Nutrition, Gazi University School of Medicine, Ankara, Turkey
| |
Collapse
|
39
|
Beyzaei Z, Ezgu F, Imanieh MH, Geramizadeh B. Identification of a novel mutation in the PHKA2 gene in a child with liver cirrhosis. J Pediatr Endocrinol Metab 2022; 35:417-420. [PMID: 34727590 DOI: 10.1515/jpem-2021-0385] [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] [Academic Contribution Register] [Received: 06/07/2021] [Accepted: 10/23/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Glycogen storage diseases (GSDs) are heterogeneous disorders caused by various enzyme deficiencies. GSD type IX α2, the most common subtype of GSD IX, is due to a deficiency of hepatic phosphorylase kinase. Herein we will report a novel mutation in this disease with an unusual presentation. CASE PRESENTATION we describe a 3-year-old boy who suffered from hepatomegaly, fatty liver disease, and liver cirrhosis. The cause of cirrhosis at a young age was unknown based on the laboratory data and liver biopsy, so we performed a targeted-gene sequencing (TGS) covering 450 genes involved in inborn metabolic diseases consisting of glycogen storage disorders genes with hepatic involvement. He was found out to have a rare novel pathogenic variant in the PHKA2 gene. CONCLUSIONS This novel variant c.2226+2T > C expands the mutational spectrum of the PHKA2 gene. Also, severe liver damage (cirrhosis) in patients with GSD- IX α2 has rarely been reported, which needs further discussion. We hypothesize that unidentified PHKA2 variants may be a rare cause of childhood liver cirrhosis.
Collapse
Affiliation(s)
- Zahra Beyzaei
- Shiraz Transplant Research Center (STRC), Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatih Ezgu
- Department of Pediatric Metabolism and Genetics, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Mohammad Hadi Imanieh
- Gastroenterology and Hepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bita Geramizadeh
- Shiraz Transplant Research Center (STRC), Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Pathology, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
40
|
Luo X, Duan Y, Fang D, Sun Y, Xiao B, Zhang H, Han L, Liang L, Gong Z, Gu X, Yu Y, Qiu W. Diagnosis and follow-up of Glycogen Storage Disease (GSD) Type VI from the largest GSD center in China. Hum Mutat 2022; 43:557-567. [PMID: 35143115 DOI: 10.1002/humu.24345] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/25/2021] [Revised: 02/03/2022] [Accepted: 02/08/2022] [Indexed: 11/10/2022]
Abstract
Glycogen storage disease (GSD) type VI is a glycogenolysis disorder caused by variants of PYGL. Knowledge about this disease is limited because only approximately 50 cases have been reported. we investigated the clinical profiles, molecular diagnosis, and treatment outcomes in patients with gsd VI from 2000 to 2021. The main initial clinical features of this cohort include hepatomegaly, short stature, elevated liver transaminases, hypertriglyceridemia, fasting hypoglycemia, and hyperuricemia. After uncooked cornstarch treatment, the stature and biochemical parameters improved significantly (P < 0.05). However, hyperuricemia recurred in most patients during adolescence. Among the 56 GSD VI patients, 54 biallelic variants and two single allelic variants of PYGL were identified, of which 43 were novel. There were two hotspot variants, c.1621-258_2178-23del and c.2467C>T p.(Gln823*), mainly in patients from Southwest and South China. c.1621-258_2178-23del is a 3.6 kb deletion that results in an out-of-frame deletion r.1621_2177del and an in-frame deletion r.1621_2265del. Our data show for the first time that long-term monitoring of uric acid is recommended for older GSD VI patients. This study also broadens the variant spectrum of PYGL and indicates that there are two hot-spot variants in China. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Xiaomei Luo
- Department of Pediatric Endocrinology and Genetics, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai.,Shanghai Institute for Pediatric Research, Shanghai, China
| | - Ying Duan
- Department of Pediatric Endocrinology and Genetics, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai
| | - Di Fang
- Department of Pediatric Endocrinology and Genetics, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai
| | - Yu Sun
- Department of Pediatric Endocrinology and Genetics, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai.,Shanghai Institute for Pediatric Research, Shanghai, China
| | - Bing Xiao
- Department of Pediatric Endocrinology and Genetics, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai.,Shanghai Institute for Pediatric Research, Shanghai, China
| | - Huiwen Zhang
- Department of Pediatric Endocrinology and Genetics, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai.,Shanghai Institute for Pediatric Research, Shanghai, China
| | - Lianshu Han
- Department of Pediatric Endocrinology and Genetics, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai.,Shanghai Institute for Pediatric Research, Shanghai, China
| | - Lili Liang
- Department of Pediatric Endocrinology and Genetics, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai.,Shanghai Institute for Pediatric Research, Shanghai, China
| | - Zhuwen Gong
- Department of Pediatric Endocrinology and Genetics, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai.,Shanghai Institute for Pediatric Research, Shanghai, China
| | - Xuefan Gu
- Department of Pediatric Endocrinology and Genetics, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai.,Shanghai Institute for Pediatric Research, Shanghai, China
| | - Yongguo Yu
- Department of Pediatric Endocrinology and Genetics, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai.,Shanghai Institute for Pediatric Research, Shanghai, China
| | - Wenjuan Qiu
- Department of Pediatric Endocrinology and Genetics, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai.,Shanghai Institute for Pediatric Research, Shanghai, China
| |
Collapse
|
41
|
Zhou Z, Su X, Cai Y, Ting TH, Zhang W, Lin Y, Xu A, Mao X, Zeng C, Liu L, Li X. Features of chinese patients with sitosterolemia. Lipids Health Dis 2022; 21:11. [PMID: 35042526 PMCID: PMC8764812 DOI: 10.1186/s12944-021-01619-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/14/2021] [Accepted: 12/24/2021] [Indexed: 11/10/2022] Open
Abstract
Background Sitosterolemia is a lipid disorder characterized by the accumulation of phytosterols in plasma and organs, caused by mutations in the ABCG5 and/or ABCG8 genes. The disease is frequently misdiagnosed and mistreated as familial hypercholesterolemia (FH). To gain a better understanding of the disease, the current status of diagnosis and treatment of Chinese patients with sitosterolemia was reviewed and summarized. Method Literature search was performed. The clinical features and molecular characteristics of Chinese patients with sitosterolemia were analysed. Four children with sitosterolemia and the treatment experience were described. Results Fifty-five patients with sitosterolemia have been reported in China. These patients were aged from 3 months to 67 years at diagnosis, and the median was 8 years of age. Several complications, such as xanthomas in 47 patients (85%), thrombocytopenia in 17 patients (31%), anemia in 14 patients (25%), and cardiovascular damage in 12 patients (22%), were observed. Thirty-nine patients (71%) exhibited mutations in the ABCG5 gene, 15 patients (27%) showed mutations in ABCG8, and variations in both genes occurred in one patient (2%). A patient with two clinically rare diseases, namely, sitosterolemia and glycogen storage disease type VI (GSD VI)), is reported here for the first time. The four reported patients were treated with low cholesterol and phytosterol-limited diet alone or combined with cholestyramine. Even though decreases were observed for total plasma cholesterol (TC) and low-density-lipoprotein cholesterol (LDL-C), and these levels were as low as normal in some patients, the levels of plant sterols remained above the normal range. However, TC, LDL-C and plant sterol levels remained at high levels in patients treated with a control diet control only. Conclusions The analysis reveals that different from Caucasians carrying mainly variations in ABCG8, most Chinese patients have mutations in the ABCG5 gene, and Arg446Ter, Gln251Ter, anArg389His might be hot-spot mutations in Chinese patients. The current survey provides clinical data to enable the development of a standardized protocol for the diagnosis and treatment of sitosterolemia in China.
Collapse
Affiliation(s)
- Zhizi Zhou
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, 510623, Guangzhou, Guangdong, China
| | - Xueying Su
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, 510623, Guangzhou, Guangdong, China
| | - Yanna Cai
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, 510623, Guangzhou, Guangdong, China
| | - Tzer Hwu Ting
- Department of Pediatrics, Faculty of Medicine & Health Sciences, University Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Wen Zhang
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, 510623, Guangzhou, Guangdong, China
| | - Yunting Lin
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, 510623, Guangzhou, Guangdong, China
| | - Aijing Xu
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, 510623, Guangzhou, Guangdong, China
| | - Xiaojian Mao
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, 510623, Guangzhou, Guangdong, China
| | - Chunhua Zeng
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, 510623, Guangzhou, Guangdong, China
| | - Li Liu
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, 510623, Guangzhou, Guangdong, China.
| | - Xiuzhen Li
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, 510623, Guangzhou, Guangdong, China.
| |
Collapse
|
42
|
Venema A, Peeks F, de Bruijn‐van der Veen M, de Boer F, Fokkert‐Wilts MJ, Lubout CMA, Huskens B, Dumont E, Mulkens S, Derks TGJ. A retrospective study of eating and psychosocial problems in patients with hepatic glycogen storage diseases and idiopathic ketotic hypoglycemia: Towards a standard set of patient-reported outcome measures. JIMD Rep 2022; 63:29-40. [PMID: 35028269 PMCID: PMC8743343 DOI: 10.1002/jmd2.12253] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 06/07/2021] [Revised: 09/06/2021] [Accepted: 09/29/2021] [Indexed: 02/05/2023] Open
Abstract
There is a paucity in literature on eating and psychosocial problems in patients with hepatic glycogen storage disease (GSD) and idiopathic ketotic hypoglycemia (IKH), problems that can greatly affect quality of life. This is a monocentre, retrospective, observational mixed method study of patients with hepatic GSD or IKH treated at the Beatrix Children's Hospital Groningen, who had been referred to SeysCentra, a specialist centre for the treatment of eating problems. Additionally, a systematic literature review has been performed to identify instruments to quantify patient-reported outcome measures of psychosocial problems in hepatic GSD patients. Sixteen patients from 12 families were included with ages ranging between 3 and 24 years. Five out of sixteen patients were diagnosed with Avoidant/Restrictive Food Intake Disorder and six patients showed characteristics of this disorder. Fourteen patients experienced sleeping problems, and 11 out of 12 parent couples experienced stress about the illness of their child. We subsequently identified 26 instruments to quantify patient-reported outcome measures for GSD patients. This study demonstrates that GSD patients can develop Avoidant/Restrictive Food Intake Disorder influencing quality of life at multiple domains. The identification of instruments to assess psychosocial wellbeing is an important step towards a standard set of patient-reported outcome measures.
Collapse
Affiliation(s)
- Annieke Venema
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Centre GroningenUniversity of GroningenGroningenThe Netherlands
| | - Fabian Peeks
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Centre GroningenUniversity of GroningenGroningenThe Netherlands
| | - Marlies de Bruijn‐van der Veen
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Centre GroningenUniversity of GroningenGroningenThe Netherlands
| | - Foekje de Boer
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Centre GroningenUniversity of GroningenGroningenThe Netherlands
| | - Marieke J. Fokkert‐Wilts
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Centre GroningenUniversity of GroningenGroningenThe Netherlands
| | - Charlotte M. A. Lubout
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Centre GroningenUniversity of GroningenGroningenThe Netherlands
| | - Bibi Huskens
- SeysCentra, Center for Paediatric Eating Problems and IncontinenceMaldenThe Netherlands
| | - Eric Dumont
- SeysCentra, Center for Paediatric Eating Problems and IncontinenceMaldenThe Netherlands
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine, and Life SciencesMaastricht UniversityMaastrichtThe Netherlands
- Department of Clinical Psychological Science, Faculty of Psychology and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
| | - Sandra Mulkens
- SeysCentra, Center for Paediatric Eating Problems and IncontinenceMaldenThe Netherlands
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine, and Life SciencesMaastricht UniversityMaastrichtThe Netherlands
- Department of Clinical Psychological Science, Faculty of Psychology and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
| | - Terry G. J. Derks
- Department of Metabolic Diseases, Beatrix Children's Hospital, University Medical Centre GroningenUniversity of GroningenGroningenThe Netherlands
| |
Collapse
|
43
|
Evaluation of Glycogen Storage Patients: Report of Twelve Novel Variants and New Clinical Findings in a Turkish Population. Genes (Basel) 2021; 12:genes12121987. [PMID: 34946936 PMCID: PMC8701369 DOI: 10.3390/genes12121987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/03/2021] [Revised: 11/26/2021] [Accepted: 12/07/2021] [Indexed: 11/17/2022] Open
Abstract
Glycogen storage diseases (GSDs) are clinically and genetically heterogeneous disorders that disturb glycogen synthesis or utilization. Although it is one of the oldest inherited metabolic disorders, new genetic methods and long-time patient follow-ups provide us with unique insight into the genotype-phenotype correlations. The aim of this study was to share the phenotypic features and molecular diagnostic results that include new pathogenic variants in our GSD cases. Twenty-six GSD patients were evaluated retrospectively. Demographic data, initial laboratory and imaging features, and current findings of the patients were recorded. Molecular analysis results were classified as novel or previously defined variants. Novel variants were analyzed with pathogenicity prediction tools according to American College of Medical Genetics and Genomics (ACGM) criteria. Twelve novel and rare variants in six different genes were associated with the disease. Hearing impairment in two patients with GSD I, early peripheral neuropathy after liver transplantation in one patient with GSD IV, epilepsy and neuromotor retardation in three patients with GSD IXA were determined. We characterized a heterogeneous group of all diagnosed GSDs over a 5-year period in our institution, and identified novel variants and new clinical findings. It is still difficult to establish a genotype-phenotype correlation in GSDs.
Collapse
|
44
|
Xiong X, Liu X, Zhu X, Tan Y, Wang Z, Xu J, Tu X, Rao Y, Duan J, Zhao W, Zhou M. A mutation in PHKG1 causes high drip loss and low meat quality in Chinese Ningdu yellow chickens. Poult Sci 2021; 101:101556. [PMID: 34852315 PMCID: PMC8639467 DOI: 10.1016/j.psj.2021.101556] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/22/2021] [Revised: 08/29/2021] [Accepted: 10/14/2021] [Indexed: 12/18/2022] Open
Abstract
With increasing societal development and the concurrent improvement in people's quality of life, meat consumption has gradually changed from a focus on “quantity” to “quality”. Broiler production is increasingly used as a means to improve meat quality by altering various characteristics, especially its genetic factors. However, until now, little has been known about the genetic variants related to meat quality traits in Chinese purebred chicken populations. To better understand these genetic underpinnings, a total of 17 traits related to meat quality and carcass were measured in 325 Chinese Ningdu yellow chickens. We performed DNA sequencing to detect nucleotide mutations, after which we conducted association studies between PHKG1 gene polymorphisms and traits related to meat quality and carcass. Results indicated a large phenotypic variation in meat quality traits. More specifically, the single nucleotide polymorphism (SNP) rs15845448 was significantly associated with drip loss at 24 h (P = 8.04 × 10−6) and 48 h (P = 5.47 × 10−6), pH (P = 2.39 × 10−3), and meat color L* (P = 9.88 × 10−3). Moreover, the SNP rs15845448 reduced 24 h and 48 h drip loss by 3.62 and 5.97%, respectively. However, no significant associations were found between rs15845448 and carcass traits (P > 0.05). Furthermore, a haplotype block containing 2 adjacent SNPs (rs15845448 and rs15845450) was identified. This block displayed 4 distinct haplotypes that had significant association with drip loss at 24 h and 48 h, pH, and meat color L*. Collectively, these results provide new insights into the genetic basis of meat quality in Chinese Ningdu yellow chickens. Moreover, the significance of SNP rs15845448 could be incorporated into the selection programs involving this breed.
Collapse
Affiliation(s)
- Xinwei Xiong
- Institute of Biological Technology, Nanchang Normal University, Nanchang, 330032, China; Key Laboratory for Genetic Improvement of Indigenous Chicken Breeds of Jiangxi Province, Nanchang, 330032, China
| | - Xianxian Liu
- Key Laboratory of Women's Reproductive Health of Jiangxi, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, 330006, China
| | - Xuenong Zhu
- Institute of Biological Technology, Nanchang Normal University, Nanchang, 330032, China; Key Laboratory for Genetic Improvement of Indigenous Chicken Breeds of Jiangxi Province, Nanchang, 330032, China
| | - Yuwen Tan
- Institute of Biological Technology, Nanchang Normal University, Nanchang, 330032, China; Key Laboratory for Genetic Improvement of Indigenous Chicken Breeds of Jiangxi Province, Nanchang, 330032, China
| | - Zhangfeng Wang
- Institute of Biological Technology, Nanchang Normal University, Nanchang, 330032, China; Key Laboratory for Genetic Improvement of Indigenous Chicken Breeds of Jiangxi Province, Nanchang, 330032, China
| | - Jiguo Xu
- Institute of Biological Technology, Nanchang Normal University, Nanchang, 330032, China; Key Laboratory for Genetic Improvement of Indigenous Chicken Breeds of Jiangxi Province, Nanchang, 330032, China
| | - Xutang Tu
- Institute of Biological Technology, Nanchang Normal University, Nanchang, 330032, China; Key Laboratory for Genetic Improvement of Indigenous Chicken Breeds of Jiangxi Province, Nanchang, 330032, China
| | - Yousheng Rao
- Institute of Biological Technology, Nanchang Normal University, Nanchang, 330032, China; Key Laboratory for Genetic Improvement of Indigenous Chicken Breeds of Jiangxi Province, Nanchang, 330032, China
| | - Jinhong Duan
- Institute of Biological Technology, Nanchang Normal University, Nanchang, 330032, China; Key Laboratory for Genetic Improvement of Indigenous Chicken Breeds of Jiangxi Province, Nanchang, 330032, China
| | - Wenliang Zhao
- Institute of Biological Technology, Nanchang Normal University, Nanchang, 330032, China; Key Laboratory for Genetic Improvement of Indigenous Chicken Breeds of Jiangxi Province, Nanchang, 330032, China
| | - Min Zhou
- Institute of Biological Technology, Nanchang Normal University, Nanchang, 330032, China; Key Laboratory for Genetic Improvement of Indigenous Chicken Breeds of Jiangxi Province, Nanchang, 330032, China.
| |
Collapse
|
45
|
Parmar K, Mosha M, Weinstein DA, Riba‐Wolman R. Method comparison of beta-hydroxybutyrate point-of-care testing to serum in healthy children. JIMD Rep 2021; 62:85-90. [PMID: 34765402 PMCID: PMC8574180 DOI: 10.1002/jmd2.12245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 05/17/2021] [Revised: 08/04/2021] [Accepted: 08/12/2021] [Indexed: 11/15/2022] Open
Abstract
Ketone production is a physiological phenomenon that occurs to avoid irreversible neurological damage from hypoglycemia, thereby serving as a marker of metabolic stress. The primary ketone body, beta-hydroxybutyrate (BHB), guides the diagnostic evaluation and management of many hypoglycemic disorders. Serum and point-of-care (POC) BHB values were not been compared in children without diabetes or metabolic disorders. We aim at comparing the serum and point-of-care BHB values in healthy children after an overnight fast. Eligible participants were ≤18 years of age prospectively recruited from elective procedures through our surgery centers. Exclusion criteria included a history of diabetes, hypopituitarism, adrenal, metabolic or inflammatory disorders, dietary restrictions, trauma, or use of medications that might affect blood glucose. The main outcome measure was comparing serum and POC BHB levels after a period of fasting. Data from 94 participants (mean age 8.29 ± 5.68 years, 54.3% male, 45.7% female, BMI mean 19.28 ± 5.25 kg/m2) were analyzed. There was a strong correlation between serum BHB (mean 0.25 ± 0.23 mmol/L) and POC BHB (mean 0.18 ± 0.20 mmol/L) (r s = 0.803, p < 0.0001). The majority (96.81%) of values for serum BHB compared with POC BHB fell within 0.1 ± 0.1 mmol/L. The average of difference between serum and POC BHB (the bias) was 0.064 mmol/L (95% CI 0.047-0.081), and percentage error was 3.19%. Point-of-care BHB is accurate and comparable to serum BHB levels in our cohort of children after an overnight fast. SYNOPSIS Point-of-care BHB agrees with serum values in healthy children.
Collapse
Affiliation(s)
- Komalben Parmar
- Department of Pediatric EndocrinologyConnecticut Children Medical CenterHartfordConnecticutUSA
- Department of PediatricsUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Maua Mosha
- Department of ResearchConnecticut Children Medical CenterHartfordConnecticutUSA
| | - David A. Weinstein
- Department of PediatricsUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Rebecca Riba‐Wolman
- Department of Pediatric EndocrinologyConnecticut Children Medical CenterHartfordConnecticutUSA
- Department of PediatricsUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| |
Collapse
|
46
|
Rossi A, Hoogeveen IJ, Lubout CMA, de Boer F, Fokkert‐Wilts MJ, Rodenburg IL, van Dam E, Grünert SC, Martinelli D, Scarpa M, Dekker H, te Boekhorst ST, van Spronsen FJ, Derks TGJ. A generic emergency protocol for patients with inborn errors of metabolism causing fasting intolerance: A retrospective, single-center study and the generation of www.emergencyprotocol.net. J Inherit Metab Dis 2021; 44:1124-1135. [PMID: 33844307 PMCID: PMC8518720 DOI: 10.1002/jimd.12386] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 12/24/2020] [Revised: 03/17/2021] [Accepted: 04/09/2021] [Indexed: 01/03/2023]
Abstract
Patients with inborn errors of metabolism causing fasting intolerance can experience acute metabolic decompensations. Long-term data on outcomes using emergency letters are lacking. This is a retrospective, observational, single-center study of the use of emergency letters based on a generic emergency protocol in patients with hepatic glycogen storage diseases (GSD) or fatty acid oxidation disorders (FAOD). Data on hospital admissions, initial laboratory results, and serious adverse events were collected. Subsequently, the website www.emergencyprotocol.net was generated in the context of the CONNECT MetabERN eHealth project following multiple meetings, protocol revisions, and translations. Representing 470 emergency protocol years, 127 hospital admissions were documented in 54/128 (42%) patients who made use of emergency letters generated based on the generic emergency protocol. Hypoglycemia (here defined as glucose concentration < 3.9 mmol/L) was reported in only 15% of hospital admissions and was uncommon in patients with ketotic GSD and patients with FAOD aged >5 years. Convulsions, coma, or death was not documented. By providing basic information, emergency letters for individual patients with hepatic GSD or the main FAOD can be generated at www.emergencyprotocol.net, in nine different languages. Generic emergency protocols are safe and easy for home management by the caregivers and the first hour in-hospital management to prevent metabolic emergencies in patients with hepatic GSD and medium-chain Acyl CoA dehydrogenase deficiency. The website www.emergencyprotocol.net is designed to support families and healthcare providers to generate personalized emergency letters for patients with hepatic GSD and the main FAOD.
Collapse
Affiliation(s)
- Alessandro Rossi
- Section of Metabolic DiseasesBeatrix Children's Hospital, University Medical Centre Groningen, University of GroningenGroningenThe Netherlands
- Department of Translational Medicine, Section of PediatricsUniversity of Naples "Federico II"NaplesItaly
| | - Irene J. Hoogeveen
- Section of Metabolic DiseasesBeatrix Children's Hospital, University Medical Centre Groningen, University of GroningenGroningenThe Netherlands
| | - Charlotte M. A. Lubout
- Section of Metabolic DiseasesBeatrix Children's Hospital, University Medical Centre Groningen, University of GroningenGroningenThe Netherlands
| | - Foekje de Boer
- Section of Metabolic DiseasesBeatrix Children's Hospital, University Medical Centre Groningen, University of GroningenGroningenThe Netherlands
| | - Marieke J. Fokkert‐Wilts
- Section of Metabolic DiseasesBeatrix Children's Hospital, University Medical Centre Groningen, University of GroningenGroningenThe Netherlands
| | - Iris L. Rodenburg
- Section of Metabolic DiseasesBeatrix Children's Hospital, University Medical Centre Groningen, University of GroningenGroningenThe Netherlands
| | - Esther van Dam
- Section of Metabolic DiseasesBeatrix Children's Hospital, University Medical Centre Groningen, University of GroningenGroningenThe Netherlands
| | - Sarah C. Grünert
- Department of General Pediatrics, Adolescent Medicine and NeonatologyMedical Centre‐University of Freiburg, Faculty of MedicineFreiburgGermany
| | - Diego Martinelli
- Division of Metabolism, Bambino Gesù Children's HospitalIRCCSRomeItaly
| | - Maurizio Scarpa
- metabERN, Regional Center for Rare DiseasesUniversity Hospital of UdineUdineItaly
| | | | - Hanka Dekker
- VKS, The Dutch Patient Association for Inherited Metabolic DiseasesZwolleThe Netherlands
| | | | - Francjan J. van Spronsen
- Section of Metabolic DiseasesBeatrix Children's Hospital, University Medical Centre Groningen, University of GroningenGroningenThe Netherlands
| | - Terry G. J. Derks
- Section of Metabolic DiseasesBeatrix Children's Hospital, University Medical Centre Groningen, University of GroningenGroningenThe Netherlands
| |
Collapse
|
47
|
Peeks F, Hoogeveen IJ, Feldbrugge RL, Burghard R, de Boer F, Fokkert‐Wilts MJ, van der Klauw MM, Oosterveer MH, Derks TGJ. A retrospective in-depth analysis of continuous glucose monitoring datasets for patients with hepatic glycogen storage disease: Recommended outcome parameters for glucose management. J Inherit Metab Dis 2021; 44:1136-1150. [PMID: 33834518 PMCID: PMC8519135 DOI: 10.1002/jimd.12383] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 01/27/2021] [Revised: 03/15/2021] [Accepted: 04/07/2021] [Indexed: 11/12/2022]
Abstract
Continuous glucose monitoring (CGM) systems have great potential for real-time assessment of glycemic variation in patients with hepatic glycogen storage disease (GSD). However, detailed descriptions and in-depth analysis of CGM data from hepatic GSD patients during interventions are scarce. This is a retrospective in-depth analysis of CGM parameters, acquired in a continuous, real-time fashion describing glucose management in 15 individual GSD patients. CGM subsets are obtained both in-hospital and at home, upon nocturnal dietary intervention (n = 1), starch loads (n = 11) and treatment of GSD Ib patients with empagliflozin (n = 3). Descriptive CGM parameters, and parameters reflecting glycemic variation and glycemic control are considered useful CGM outcome parameters. Furthermore, the combination of first and second order derivatives, cumulative sum and Fourier analysis identified both subtle and sudden changes in glucose management; hence, aiding assessment of dietary and medical interventions. CGM data interpolation for nocturnal intervals reduced confounding by physical activity and diet. Based on these analyses, we conclude that in-depth CGM analysis can be a powerful tool to assess glucose management and optimize treatment in individual hepatic GSD patients.
Collapse
Affiliation(s)
- Fabian Peeks
- Section of Metabolic DiseasesBeatrix Children's Hospital, University Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - Irene J. Hoogeveen
- Section of Metabolic DiseasesBeatrix Children's Hospital, University Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | | | | | - Foekje de Boer
- Section of Metabolic DiseasesBeatrix Children's Hospital, University Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - Marieke J. Fokkert‐Wilts
- Section of Metabolic DiseasesBeatrix Children's Hospital, University Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - Melanie M. van der Klauw
- Department of EndocrinologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Maaike H. Oosterveer
- Laboratory of PediatricsUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Terry G. J. Derks
- Section of Metabolic DiseasesBeatrix Children's Hospital, University Medical Center Groningen, University of GroningenGroningenThe Netherlands
| |
Collapse
|
48
|
Bone Mineral Density in Patients with Hepatic Glycogen Storage Diseases. Nutrients 2021; 13:nu13092987. [PMID: 34578865 PMCID: PMC8469033 DOI: 10.3390/nu13092987] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/10/2020] [Revised: 01/18/2021] [Accepted: 02/06/2021] [Indexed: 01/10/2023] Open
Abstract
The association between bone mineral density (BMD) and hepatic glycogen storage diseases (GSDs) is still unclear. To evaluate the BMD of patients with GSD I, IIIa and IXα, a cross-sectional study was performed, including 23 patients (GSD Ia = 13, Ib = 5, IIIa = 2 and IXα = 3; median age = 11.9 years; IQ = 10.9–20.1) who underwent a dual-energy X-ray absorptiometry (DXA). Osteocalcin (OC, n = 18), procollagen type 1 N-terminal propeptide (P1NP, n = 19), collagen type 1 C-terminal telopeptide (CTX, n = 18) and 25-OH Vitamin D (n = 23) were also measured. The participants completed a 3-day food diary (n = 20). Low BMD was defined as a Z-score ≤ −2.0. All participants were receiving uncooked cornstarch (median dosage = 6.3 g/kg/day) at inclusion, and 11 (47.8%) presented good metabolic control. Three (13%) patients (GSD Ia = 1, with poor metabolic control; IIIa = 2, both with high CPK levels) had a BMD ≤ −2.0. CTX, OC and P1NP correlated negatively with body weight and age. 25-OH Vitamin D concentration was decreased in seven (30.4%) patients. Our data suggest that patients with hepatic GSDs may have low BMD, especially in the presence of muscular involvement and poor metabolic control. Systematic nutritional monitoring of these patients is essential.
Collapse
|
49
|
Marciano CMM, Ibelli AMG, Marchesi JAP, de Oliveira Peixoto J, Fernandes LT, Savoldi IR, do Carmo KB, Ledur MC. Differential Expression of Myogenic and Calcium Signaling-Related Genes in Broilers Affected With White Striping. Front Physiol 2021; 12:712464. [PMID: 34381378 PMCID: PMC8349984 DOI: 10.3389/fphys.2021.712464] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/20/2021] [Accepted: 07/05/2021] [Indexed: 01/10/2023] Open
Abstract
White Striping (WS) has been one of the main issues in poultry production in the last years since it affects meat quality. Studies have been conducted to understand WS and other myopathies in chickens, and some biological pathways have been associated to the prevalence of these conditions, such as extracellular calcium level, oxidative stress, localized hypoxia, possible fiber-type switching, and cellular repairing. Therefore, to understand the genetic mechanisms involved in WS, 15 functional candidate genes were chosen to be analyzed by quantitative PCR (qPCR) in breast muscle of normal and WS-affected chickens. To this, the pectoral major muscle (PMM) of 16 normal and 16 WS-affected broilers were collected at 42 days of age and submitted to qRT-PCR analysis. Out of the 15 genes studied, six were differentially expressed between groups. The CA2, CSRP3, and PLIN1 were upregulated, while CALM2, DNASE1L3, and MYLK2 genes were downregulated in the WS-affected when compared to the normal broilers. These findings highlight that the disruption on muscle and calcium signaling pathways can possibly be triggering WS in chickens. Improving our understanding on the genetic basis involved with this myopathy might contribute for reducing WS in poultry production.
Collapse
Affiliation(s)
| | - Adriana Mércia Guaratini Ibelli
- Embrapa Suínos e Aves, Concórdia, Brazil.,Programa de Pós-Graduação em Ciências Veterinárias, Universidade Estadual do Centro-Oeste, Guarapuava, Brazil
| | | | - Jane de Oliveira Peixoto
- Embrapa Suínos e Aves, Concórdia, Brazil.,Programa de Pós-Graduação em Ciências Veterinárias, Universidade Estadual do Centro-Oeste, Guarapuava, Brazil
| | | | - Igor Ricardo Savoldi
- Programa de Pós-Graduação em Zootecnia, Universidade do Estado de Santa Catarina (UDESC-Oeste), Chapecó, Brazil
| | | | - Mônica Corrêa Ledur
- Programa de Pós-Graduação em Zootecnia, Universidade do Estado de Santa Catarina (UDESC-Oeste), Chapecó, Brazil.,Embrapa Suínos e Aves, Concórdia, Brazil
| |
Collapse
|
50
|
Gibson RA, Lim JA, Choi SJ, Flores L, Clinton L, Bali D, Young S, Asokan A, Sun B, Kishnani PS. Characterization of liver GSD IX γ2 pathophysiology in a novel Phkg2 -/- mouse model. Mol Genet Metab 2021; 133:269-276. [PMID: 34083142 PMCID: PMC9792075 DOI: 10.1016/j.ymgme.2021.05.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 12/04/2020] [Revised: 05/15/2021] [Accepted: 05/22/2021] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Liver Glycogen Storage Disease IX is a rare metabolic disorder of glycogen metabolism caused by deficiency of the phosphorylase kinase enzyme (PhK). Variants in the PHKG2 gene, encoding the liver-specific catalytic γ2 subunit of PhK, are associated with a liver GSD IX subtype known as PHKG2 GSD IX or GSD IX γ2. There is emerging evidence that patients with GSD IX γ2 can develop severe and progressive liver disease, yet research regarding the disease has been minimal to date. Here we characterize the first mouse model of liver GSD IX γ2. METHODS A Phkg2-/- mouse model was generated via targeted removal of the Phkg2 gene. Knockout (Phkg2-/-, KO) and wild type (Phkg2+/+, WT) mice up to 3 months of age were compared for morphology, Phkg2 transcription, PhK enzyme activity, glycogen content, histology, serum liver markers, and urinary glucose tetrasaccharide Glcα1-6Glcα1-4Glcα1-4Glc (Glc4). RESULTS When compared to WT controls, KO mice demonstrated significantly decreased liver PhK enzyme activity, increased liver: body weight ratio, and increased glycogen in the liver, with no glycogen accumulation observed in the brain, quadricep, kidney, and heart. KO mice demonstrated elevated liver blood markers as well as elevated urine Glc4, a commonly used biomarker for glycogen storage disease. KO mice demonstrated features of liver structural damage. Hematoxylin & Eosin and Masson's Trichrome stained KO mice liver histology slides revealed characteristic GSD hepatocyte architectural changes and early liver fibrosis, as have been reported in liver GSD patients. DISCUSSION This study provides the first evidence of a mouse model that recapitulates the liver-specific pathology of patients with GSD IX γ2. The model will provide the first platform for further study of disease progression in GSD IX γ2 as well as for the evaluation of novel therapeutics.
Collapse
Affiliation(s)
- Rebecca A Gibson
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA; Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Jeong-A Lim
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Su Jin Choi
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Leticia Flores
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Lani Clinton
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Deeksha Bali
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Sarah Young
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Aravind Asokan
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA; Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Baodong Sun
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Priya S Kishnani
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA; Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|