1
|
Monteillet L, Perrot G, Evrard F, Miliano A, Silva M, Leblond A, Nguyen C, Terzi F, Mithieux G, Rajas F. Impaired Glucose Metabolism, Primary Cilium Defects, and Kidney Cystogenesis in Glycogen Storage Disease Type Ia. J Am Soc Nephrol 2024:00001751-990000000-00394. [PMID: 39141438 DOI: 10.1681/asn.0000000000000452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 07/31/2024] [Indexed: 08/16/2024] Open
Abstract
Key Points
Metabolism adaptations due to glucose-6 phosphate accumulation in glycogen storage disease type Ia kidneys, toward a Warburg-like metabolism, promoted cell proliferation.Metabolic perturbations directly affected primary cilium structure and cystogenesis in glycogen storage disease type Ia kidneys.
Background
Glycogen storage disease type Ia (GSDIa) is a rare metabolic disorder caused by mutations in the catalytic subunit of glucose-6 phosphatase (G6PC1). This leads to severe hypoglycemia, and most young patients with GSDIa develop CKD. The kidney pathology is characterized by the development of cysts, which typically occur at an advanced stage of CKD.
Methods
To elucidate the molecular mechanisms responsible for cyst formation, we characterized renal metabolism, molecular pathways involved in cell proliferation, and primary cilium integrity using mice in which G6pc1 was specifically deleted in the kidney from an in utero stage.
Results
GSDIa mice exhibited kidney fibrosis, high inflammation, and cyst formation, leading to kidney dysfunction. In addition, the loss of G6PC1 led to the ectopic accumulation of glycogen and lipids in the kidneys and a metabolic shift toward a Warburg-like metabolism. This metabolic adaptation was due to an excess of glucose-6 phosphate, which supports cell proliferation, driven by the mitogen-activated protein kinase/extracellular signal–regulated kinases and protein kinase B/mammalian target of rapamycin pathways. Treatment of GSDIa mice with rapamycin, a target of the mammalian target of rapamycin pathway, reduced cell proliferation and kidney damage. Our results also identified lipocalin 2 as a contributor to renal inflammation and an early biomarker of CKD progression in GSDIa mice. Its inactivation partially prevented kidney lesions in GSDIa. Importantly, primary cilium defects were observed in the kidneys of GSDIa mice.
Conclusions
Metabolic adaptations because of glucose-6 phosphate accumulation in GSDIa renal tubules, toward a Warburg-like metabolism, promoted cell proliferation and cyst formation in a similar manner to that observed in various cystic kidney diseases. This was associated with downregulation of primary cilium gene expression and, consequently, altered cilium morphology.
Collapse
Affiliation(s)
- Laure Monteillet
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| | - Gwendoline Perrot
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| | - Félicie Evrard
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| | - Alexane Miliano
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| | - Marine Silva
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| | - Alicia Leblond
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| | - Clément Nguyen
- Université de Paris Cité, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades, Département "Croissance et Signalisation," Paris, France
| | - Fabiola Terzi
- Université de Paris Cité, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades, Département "Croissance et Signalisation," Paris, France
| | - Gilles Mithieux
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| | - Fabienne Rajas
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| |
Collapse
|
2
|
Liu T, Wang L, Ji L, Mu L, Wang K, Xu G, Wang S, Ma Q. Plantaginis Semen Ameliorates Hyperuricemia Induced by Potassium Oxonate. Int J Mol Sci 2024; 25:8548. [PMID: 39126116 PMCID: PMC11313179 DOI: 10.3390/ijms25158548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
Plantaginis semen is the dried ripe seed of Plantago asiatica L. or Plantago depressa Willd., which has a long history in alleviating hyperuricemia (HUA) and chronic kidney diseases. While the major chemical ingredients and mechanism remained to be illustrated. Therefore, this work aimed to elucidate the chemicals and working mechanisms of PS for HUA. UPLC-QE-Orbitrap-MS was applied to identify the main components of PS in vitro and in vivo. RNA sequencing (RNA-seq) was conducted to explore the gene expression profile, and the genes involved were further confirmed by real-time quantitative PCR (RT-qPCR). A total of 39 components were identified from PS, and 13 of them were detected in the rat serum after treating the rat with PS. The kidney tissue injury and serum uric acid (UA), xanthine oxidase (XOD), and cytokine levels were reversed by PS. Meanwhile, renal urate anion transporter 1 (Urat1) and glucose transporter 9 (Glut9) levels were reversed with PS treatment. RNA-seq analysis showed that the PPAR signaling pathway; glycine, serine, and threonine metabolism signaling pathway; and fatty acid metabolism signaling pathway were significantly modified by PS treatment. Further, the gene expression of Slc7a8, Pck1, Mgll, and Bhmt were significantly elevated, and Fkbp5 was downregulated, consistent with RNA-seq results. The PPAR signaling pathway involved Pparα, Pparγ, Lpl, Plin5, Atgl, and Hsl were elevated by PS treatment. URAT1 and PPARα proteins levels were confirmed by Western blotting. In conclusion, this study elucidates the chemical profile and working mechanisms of PS for prevention and therapy of HUA and provides a promising traditional Chinese medicine agency for HUA prophylaxis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shifeng Wang
- Key Laboratory of TCM-Information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; (T.L.); (L.W.); (L.J.); (L.M.); (K.W.); (G.X.)
| | - Qun Ma
- Key Laboratory of TCM-Information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; (T.L.); (L.W.); (L.J.); (L.M.); (K.W.); (G.X.)
| |
Collapse
|
3
|
Hatano R, Lee E, Sato H, Kiuchi M, Hirahara K, Nakagawa Y, Shimano H, Nakayama T, Tanaka T, Miki T. Hepatic ketone body regulation of renal gluconeogenesis. Mol Metab 2024; 84:101934. [PMID: 38604598 PMCID: PMC11039402 DOI: 10.1016/j.molmet.2024.101934] [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] [Scholar Register] [Received: 01/08/2024] [Revised: 02/20/2024] [Accepted: 04/02/2024] [Indexed: 04/13/2024] Open
Abstract
OBJECTIVES During fasting, liver pivotally regulates blood glucose levels through glycogenolysis and gluconeogenesis. Kidney also produces glucose through gluconeogenesis. Gluconeogenic genes are transactivated by fasting, but their expression patterns are chronologically different between the two organs. We find that renal gluconeogenic gene expressions are positively correlated with the blood β-hydroxybutyrate concentration. Thus, we herein aim to investigate the regulatory mechanism and its physiological implications. METHODS Gluconeogenic gene expressions in liver and kidney were examined in hyperketogenic mice such as high-fat diet (HFD)-fed and ketogenic diet-fed mice, and in hypoketogenic PPARα knockout (PPARα-/-) mice. Renal gluconeogenesis was evaluated by rise in glycemia after glutamine loading in vivo. Functional roles of β-hydroxybutyrate in the regulation of renal gluconeogenesis were investigated by metabolome analysis and RNA-seq analysis of proximal tubule cells. RESULTS Renal gluconeogenic genes were transactivated concurrently with blood β-hydroxybutyrate uprise under ketogenic states, but the increase was blunted in hypoketogenic PPARα-/- mice. Administration of 1,3-butandiol, a ketone diester, transactivated renal gluconeogenic gene expression in fasted PPARα-/- mice. In addition, HFD-fed mice showed fasting hyperglycemia along with upregulated renal gluconeogenic gene expression, which was blunted in HFD-fed PPARα-/- mice. In vitro experiments and metabolome analysis in renal tubular cells showed that β-hydroxybutyrate directly promotes glucose and NH3 production through transactivating gluconeogenic genes. In addition, RNA-seq analysis revealed that β-hydroxybutyrate-induced transactivation of Pck1 was mediated by C/EBPβ. CONCLUSIONS Our findings demonstrate that β-hydroxybutyrate mediates hepato-renal interaction to maintain homeostatic regulation of blood glucose and systemic acid-base balance through renal gluconeogenesis regulation.
Collapse
Affiliation(s)
- Ryo Hatano
- Department of Medical Physiology, Chiba University, Graduate School of Medicine, Chiba 260-8670, Japan
| | - Eunyoung Lee
- Department of Medical Physiology, Chiba University, Graduate School of Medicine, Chiba 260-8670, Japan; Research Institute of Disaster Medicine (RIDM), Chiba University, Graduate School of Medicine, Chiba 260-8670, Japan
| | - Hiromi Sato
- Laboratory of Clinical Pharmacology and Pharmacometrics, Chiba University, Graduate School of Pharmaceutical Sciences, Chiba 260-8670, Japan
| | - Masahiro Kiuchi
- Department of Immunology, Chiba University, Graduate School of Medicine, Chiba 260-8670, Japan
| | - Kiyoshi Hirahara
- Research Institute of Disaster Medicine (RIDM), Chiba University, Graduate School of Medicine, Chiba 260-8670, Japan; Department of Immunology, Chiba University, Graduate School of Medicine, Chiba 260-8670, Japan
| | - Yoshimi Nakagawa
- Division of Complex Biosystem Research, Department of Research and Development, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Hitoshi Shimano
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Toshinori Nakayama
- Department of Immunology, Chiba University, Graduate School of Medicine, Chiba 260-8670, Japan
| | - Tomoaki Tanaka
- Research Institute of Disaster Medicine (RIDM), Chiba University, Graduate School of Medicine, Chiba 260-8670, Japan; Department of Molecular Diagnosis, Chiba University, Graduate School of Medicine, Chiba 260-8670, Japan
| | - Takashi Miki
- Department of Medical Physiology, Chiba University, Graduate School of Medicine, Chiba 260-8670, Japan; Research Institute of Disaster Medicine (RIDM), Chiba University, Graduate School of Medicine, Chiba 260-8670, Japan.
| |
Collapse
|
4
|
Koeberl DD, Koch RL, Lim JA, Brooks ED, Arnson BD, Sun B, Kishnani PS. Gene therapy for glycogen storage diseases. J Inherit Metab Dis 2024; 47:93-118. [PMID: 37421310 PMCID: PMC10874648 DOI: 10.1002/jimd.12654] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 05/24/2023] [Accepted: 07/05/2023] [Indexed: 07/10/2023]
Abstract
Glycogen storage disorders (GSDs) are inherited disorders of metabolism resulting from the deficiency of individual enzymes involved in the synthesis, transport, and degradation of glycogen. This literature review summarizes the development of gene therapy for the GSDs. The abnormal accumulation of glycogen and deficiency of glucose production in GSDs lead to unique symptoms based upon the enzyme step and tissues involved, such as liver and kidney involvement associated with severe hypoglycemia during fasting and the risk of long-term complications including hepatic adenoma/carcinoma and end stage kidney disease in GSD Ia from glucose-6-phosphatase deficiency, and cardiac/skeletal/smooth muscle involvement associated with myopathy +/- cardiomyopathy and the risk for cardiorespiratory failure in Pompe disease. These symptoms are present to a variable degree in animal models for the GSDs, which have been utilized to evaluate new therapies including gene therapy and genome editing. Gene therapy for Pompe disease and GSD Ia has progressed to Phase I and Phase III clinical trials, respectively, and are evaluating the safety and bioactivity of adeno-associated virus vectors. Clinical research to understand the natural history and progression of the GSDs provides invaluable outcome measures that serve as endpoints to evaluate benefits in clinical trials. While promising, gene therapy and genome editing face challenges with regard to clinical implementation, including immune responses and toxicities that have been revealed during clinical trials of gene therapy that are underway. Gene therapy for the glycogen storage diseases is under development, addressing an unmet need for specific, stable therapy for these conditions.
Collapse
Affiliation(s)
- Dwight D Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical School, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Rebecca L Koch
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical School, Durham, North Carolina, USA
| | - Jeong-A Lim
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical School, Durham, North Carolina, USA
| | - Elizabeth D Brooks
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical School, Durham, North Carolina, USA
| | - Benjamin D Arnson
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Baodong Sun
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical School, Durham, North Carolina, USA
| | - Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical School, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
5
|
Lee C, Pratap K, Zhang L, Chen HD, Gautam S, Arnaoutova I, Raghavankutty M, Starost MF, Kahn M, Mansfield BC, Chou JY. Inhibition of Wnt/β-catenin signaling reduces renal fibrosis in murine glycogen storage disease type Ia. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166874. [PMID: 37666439 PMCID: PMC10841171 DOI: 10.1016/j.bbadis.2023.166874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 08/17/2023] [Accepted: 08/31/2023] [Indexed: 09/06/2023]
Abstract
Glycogen storage disease type Ia (GSD-Ia) is caused by a deficiency in the enzyme glucose-6-phosphatase-α (G6Pase-α or G6PC) that is expressed primarily in the gluconeogenic organs, namely liver, kidney cortex, and intestine. Renal G6Pase-α deficiency in GSD-Ia is characterized by impaired gluconeogenesis, nephromegaly due to elevated glycogen accumulation, and nephropathy caused, in part, by renal fibrosis, mediated by activation of the renin-angiotensin system (RAS). The Wnt/β-catenin signaling regulates the expression of a variety of downstream mediators implicated in renal fibrosis, including multiple genes in the RAS. Sustained activation of Wnt/β-catenin signaling is associated with the development and progression of renal fibrotic lesions that can lead to chronic kidney disease. In this study, we examined the molecular mechanism underlying GSD-Ia nephropathy. Damage to the kidney proximal tubules is known to trigger acute kidney injury (AKI) that can, in turn, activate Wnt/β-catenin signaling. We show that GSD-Ia mice have AKI that leads to activation of the Wnt/β-catenin/RAS axis. Renal fibrosis was demonstrated by increased renal levels of Snail1, α-smooth muscle actin (α-SMA), and extracellular matrix proteins, including collagen-Iα1 and collagen-IV. Treating GSD-Ia mice with a CBP/β-catenin inhibitor, ICG-001, significantly decreased nuclear translocated active β-catenin and reduced renal levels of renin, Snail1, α-SMA, and collagen-IV. The results suggest that inhibition of Wnt/β-catenin signaling may be a promising therapeutic strategy for GSD-Ia nephropathy.
Collapse
Affiliation(s)
- Cheol Lee
- Section on Cellular Differentiation, Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20802, USA
| | - Kunal Pratap
- Section on Cellular Differentiation, Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20802, USA
| | - Lisa Zhang
- Section on Cellular Differentiation, Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20802, USA
| | - Hung Dar Chen
- Section on Cellular Differentiation, Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20802, USA
| | - Sudeep Gautam
- Section on Cellular Differentiation, Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20802, USA
| | - Irina Arnaoutova
- Section on Cellular Differentiation, Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20802, USA
| | - Mahadevan Raghavankutty
- Section on Developmental Genetics, Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20802, USA
| | - Matthew F Starost
- Division of Veterinary Resources, National Institutes of Health, Bethesda, MD 20802, USA
| | - Michael Kahn
- Department of Cancer Biology and Molecular Medicine, Beckmann Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Brian C Mansfield
- Section on Cellular Differentiation, Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20802, USA
| | - Janice Y Chou
- Section on Cellular Differentiation, Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20802, USA.
| |
Collapse
|
6
|
Ito K, Tajima G, Kamisato C, Tsumura M, Iwamoto M, Sekiguchi Y, Numata Y, Watanabe K, Yabe Y, Kanki S, Fujieda Y, Goto K, Sogawa Y, Oitate M, Nagase H, Tsuji S, Nishizawa T, Kakuta M, Masuda T, Onishi Y, Koizumi M, Nakamura H, Okada S, Matsuo M, Takaishi K. A splice-switching oligonucleotide treatment ameliorates glycogen storage disease type 1a in mice with G6PC c.648G>T. J Clin Invest 2023; 133:e163464. [PMID: 37788110 PMCID: PMC10688987 DOI: 10.1172/jci163464] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/27/2023] [Indexed: 10/05/2023] Open
Abstract
Glycogen storage disease type 1a (GSD1a) is caused by a congenital deficiency of glucose-6-phosphatase-α (G6Pase-α, encoded by G6PC), which is primarily associated with life-threatening hypoglycemia. Although strict dietary management substantially improves life expectancy, patients still experience intermittent hypoglycemia and develop hepatic complications. Emerging therapies utilizing new modalities such as adeno-associated virus and mRNA with lipid nanoparticles are under development for GSD1a but potentially require complicated glycemic management throughout life. Here, we present an oligonucleotide-based therapy to produce intact G6Pase-α from a pathogenic human variant, G6PC c.648G>T, the most prevalent variant in East Asia causing aberrant splicing of G6PC. DS-4108b, a splice-switching oligonucleotide, was designed to correct this aberrant splicing, especially in liver. We generated a mouse strain with homozygous knockin of this variant that well reflected the pathophysiology of patients with GSD1a. DS-4108b recovered hepatic G6Pase activity through splicing correction and prevented hypoglycemia and various hepatic abnormalities in the mice. Moreover, DS-4108b had long-lasting efficacy of more than 12 weeks in mice that received a single dose and had favorable pharmacokinetics and tolerability in mice and monkeys. These findings together indicate that this oligonucleotide-based therapy could provide a sustainable and curative therapeutic option under easy disease management for GSD1a patients with G6PC c.648G>T.
Collapse
Affiliation(s)
- Kentaro Ito
- Specialty Medicine Research Laboratories I, Daiichi Sankyo Co., Ltd., Tokyo, Japan
- Department of Pediatrics, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Go Tajima
- Department of Pediatrics, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
- Division of Neonatal Screening, Research Institute, National Center for Child Health and Development, Tokyo, Japan
| | - Chikako Kamisato
- Specialty Medicine Research Laboratories I, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Miyuki Tsumura
- Department of Pediatrics, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | | | | | | | - Kyoko Watanabe
- Drug Metabolism and Pharmacokinetics Research Laboratories
| | - Yoshiyuki Yabe
- Drug Metabolism and Pharmacokinetics Research Laboratories
| | - Satomi Kanki
- Drug Metabolism and Pharmacokinetics Research Laboratories
| | | | - Koichi Goto
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | | | | | - Hiroyuki Nagase
- Specialty Medicine Research Laboratories I, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Shinnosuke Tsuji
- Specialty Medicine Research Laboratories I, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Tomohiro Nishizawa
- Specialty Medicine Research Laboratories I, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Masayo Kakuta
- Specialty Medicine Research Laboratories I, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | | | | | | | - Hidefumi Nakamura
- Department of Research and Development Supervision, National Center for Child Health and Development, Tokyo, Japan
| | - Satoshi Okada
- Department of Pediatrics, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Masafumi Matsuo
- Research Center for Locomotion Biology, Kobe Gakuin University, Kobe, Japan
| | - Kiyosumi Takaishi
- Specialty Medicine Research Laboratories I, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| |
Collapse
|
7
|
Küçükçongar Yavaş A, Engin Erdal A, Bilginer Gürbüz B, Ünlüsoy Aksu A, Kasapkara ÇS. Assessment of the diagnosis, treatment, and follow-up of a group of Turkish pediatric glycogen storage disease type 1b patients with varying clinical presentations and a novel mutation. J Pediatr Endocrinol Metab 2023; 36:1092-1099. [PMID: 37791580 DOI: 10.1515/jpem-2023-0336] [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] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/18/2023] [Indexed: 10/05/2023]
Abstract
OBJECTIVES Glycogen storage disease (GSD) type 1b is a multisystemic disease in which immune and infectious complications are present, different from GSD type 1a. Treatment with granulocyte-colony stimulating factor (G-CSF) is often required in the management of neutropenia and inflammatory bowel disease. Recently, an alternative treatment option to G-CSF has been preferred, like empagliflozin. To report on the demographics, genotype, clinical presentation, management, and complications of pediatric patients with glycogen storage disease type 1b (GSD 1b). METHODS A retrospective analysis of the clinical course of eight patients with GSD type 1b whose diagnosis was confirmed by molecular testing. RESULTS The mean age at referral was four months. The diagnosis of GSD 1b was based on clinical and laboratory findings and supported by genetic studies. One patient presented with an atypical clinical finding in the form of hydrocephalus at the time of first admission. The first symptom was abscess formation on the scalp due to neutropenia in another patient. Other patients had hypoglycemia at the time of admission. All patients presented suffered from neutropenia, which was managed with G-CSF, except one. Hospitalizations for infections were frequent. One patient developed chronic diarrhea and severe infections, which have been brought under control with empagliflozin. CONCLUSIONS Neutropenia is an essential finding in GSD 1b and responsible for complications. The coexistence of hypoglycemia and neutropenia should bring to mind GSD 1b. Empagliflozin can be a treatment option for neutropenia, which is resistant to G-CSF treatment.
Collapse
Affiliation(s)
- Aynur Küçükçongar Yavaş
- Department of Pediatric Metabolic Diseases, Children's Hospital, Ankara Bilkent City Hospital, Çankaya, Ankara, Türkiye
| | - Ayşenur Engin Erdal
- Department of Pediatric Metabolic Diseases, Children's Hospital, Ankara Bilkent City Hospital, Çankaya, Ankara, Türkiye
| | - Berrak Bilginer Gürbüz
- Department of Pediatric Metabolic Diseases, Children's Hospital, Ankara Bilkent City Hospital, Çankaya, Ankara, Türkiye
| | - Aysel Ünlüsoy Aksu
- Department of Pediatric Gastroenterology Diseases, Children's Hospital, Ankara Bilkent City Hospital, Ankara, Türkiye
| | - Çiğdem Seher Kasapkara
- Department of Pediatric Metabolic Diseases, Children's Hospital, Ankara Bilkent City Hospital, Çankaya, Ankara, Türkiye
| |
Collapse
|
8
|
Zhong J, Gou Y, Zhao P, Dong X, Guo M, Li A, Hao A, Luu HH, He TC, Reid RR, Fan J. Glycogen storage disease type I: Genetic etiology, clinical manifestations, and conventional and gene therapies. PEDIATRIC DISCOVERY 2023; 1:e3. [PMID: 38370424 PMCID: PMC10874634 DOI: 10.1002/pdi3.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 05/11/2023] [Indexed: 02/20/2024]
Abstract
Glycogen storage disease type I (GSDI) is an inherited metabolic disorder characterized by a deficiency of enzymes or proteins involved in glycogenolysis and gluconeogenesis, resulting in excessive intracellular glycogen accumulation. While GSDI is classified into four different subtypes based on molecular genetic variants, GSDIa accounts for approximately 80%. GSDIa and GSDIb are autosomal recessive disorders caused by deficiencies in glucose-6-phosphatase (G6Pase-α) and glucose-6-phosphate-transporter (G6PT), respectively. For the past 50 years, the care of patients with GSDI has been improved following elaborate dietary managements. GSDI patients currently receive dietary therapies that enable patients to improve hypoglycemia and alleviate early symptomatic signs of the disease. However, dietary therapies have many limitations with a risk of calcium, vitamin D, and iron deficiency and cannot prevent long-term complications, such as progressive liver and renal failure. With the deepening understanding of the pathogenesis of GSDI and the development of gene therapy technology, there is great progress in the treatment of GSDI. Here, we review the underlying molecular genetics and the current clinical management strategies of GSDI patients with an emphasis on promising experimental gene therapies.
Collapse
Affiliation(s)
- Jiamin Zhong
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois, USA
| | - Yannian Gou
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois, USA
| | - Piao Zhao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois, USA
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiangyu Dong
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Meichun Guo
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Aohua Li
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Ailing Hao
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Hue H. Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois, USA
- Laboratory of Craniofacial Biology and Development, Department of Surgery, Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, Illinois, USA
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois, USA
- Laboratory of Craniofacial Biology and Development, Department of Surgery, Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, Illinois, USA
| | - Jiaming Fan
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois, USA
| |
Collapse
|
9
|
Su YY, Liu YL, Huang HC, Lin CC. Ensemble learning model for identifying the hallmark genes of NFκB/TNF signaling pathway in cancers. J Transl Med 2023; 21:485. [PMID: 37475016 PMCID: PMC10357720 DOI: 10.1186/s12967-023-04355-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 07/13/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND The nuclear factor kappa B (NFκB) regulatory pathways downstream of tumor necrosis factor (TNF) play a critical role in carcinogenesis. However, the widespread influence of NFκB in cells can result in off-target effects, making it a challenging therapeutic target. Ensemble learning is a machine learning technique where multiple models are combined to improve the performance and robustness of the prediction. Accordingly, an ensemble learning model could uncover more precise targets within the NFκB/TNF signaling pathway for cancer therapy. METHODS In this study, we trained an ensemble learning model on the transcriptome profiles from 16 cancer types in the TCGA database to identify a robust set of genes that are consistently associated with the NFκB/TNF pathway in cancer. Our model uses cancer patients as features to predict the genes involved in the NFκB/TNF signaling pathway and can be adapted to predict the genes for different cancer types by switching the cancer type of patients. We also performed functional analysis, survival analysis, and a case study of triple-negative breast cancer to demonstrate our model's potential in translational cancer medicine. RESULTS Our model accurately identified genes regulated by NFκB in response to TNF in cancer patients. The downstream analysis showed that the identified genes are typically involved in the canonical NFκB-regulated pathways, particularly in adaptive immunity, anti-apoptosis, and cellular response to cytokine stimuli. These genes were found to have oncogenic properties and detrimental effects on patient survival. Our model also could distinguish patients with a specific cancer subtype, triple-negative breast cancer (TNBC), which is known to be influenced by NFκB-regulated pathways downstream of TNF. Furthermore, a functional module known as mononuclear cell differentiation was identified that accurately predicts TNBC patients and poor short-term survival in non-TNBC patients, providing a potential avenue for developing precision medicine for cancer subtypes. CONCLUSIONS In conclusion, our approach enables the discovery of genes in NFκB-regulated pathways in response to TNF and their relevance to carcinogenesis. We successfully categorized these genes into functional groups, providing valuable insights for discovering more precise and targeted cancer therapeutics.
Collapse
Affiliation(s)
- Yin-Yuan Su
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Ling Liu
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of General Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Hsuan-Cheng Huang
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chen-Ching Lin
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
10
|
Lenzini L, Iori E, Scannapieco F, Carraro G, Avogaro A, Vitturi N. Urine-Derived Epithelial Cells as a New Model to Study Renal Metabolic Phenotypes of Patients with Glycogen Storage Disease 1a. Int J Mol Sci 2022; 24:ijms24010232. [PMID: 36613675 PMCID: PMC9820562 DOI: 10.3390/ijms24010232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
Glycogen storage diseases (GSDs) represent a model of pathological accumulation of glycogen disease in the kidney that, in animal models, results in nephropathy due to abnormal autophagy and mitochondrial function. Patients with Glycogen Storage Disease 1a (GSD1a) accumulate glycogen in the kidneys and suffer a disease resembling diabetic nephropathy that can progress to renal failure. In this study, we addressed whether urine-derived epithelial cells (URECs) from patients with GSD1a maintain their biological features, and whether they can be used as a model to study the renal and metabolic phenotypes of this genetic condition. Studies were performed on cells extracted from urine samples of GSD1a and healthy subjects. URECs were characterized after the fourth passage by transmission electron microscopy and immunofluorescence. Reactive oxygen species (ROS), at different glucose concentrations, were measured by fluorescent staining. We cultured URECs from three patients with GSD1a and three healthy controls. At the fourth passage, URECs from GSD1a patients maintained their massive glycogen content. GSD1a and control cells showed the ciliary structures of renal tubular epithelium and the expression of epithelial (E-cadherin) and renal tubular cells (aquaporin 1 and 2) markers. Moreover, URECs from both groups responded to changes in glucose concentrations by modulating ROS levels. GSD1a cells were featured by a specific response to the low glucose stimulus, which is the condition that more resembles the metabolic derangement of patients with GSD1a. Through this study, we demonstrated that URECs might represent a promising experimental model to study the molecular mechanisms leading to renal damage in GSD1a, due to pathological glycogen storage.
Collapse
Affiliation(s)
- Livia Lenzini
- Emergency Medicine Unit and Specialized Center of Excellence for Hypertension of the European Society of Hypertension, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
| | - Elisabetta Iori
- Division of Metabolic Diseases, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
| | - Federico Scannapieco
- Division of Metabolic Diseases, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
| | - Gianni Carraro
- Nephrology, Dialysis and Transplant Unit, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
| | - Angelo Avogaro
- Division of Metabolic Diseases, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
| | - Nicola Vitturi
- Division of Metabolic Diseases, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
- Correspondence: ; Tel.: +39-049-821-4326
| |
Collapse
|
11
|
Petrova IO, Smirnikhina SA. Studies on glycogen storage disease type 1a animal models: a brief perspective. Transgenic Res 2022; 31:593-606. [PMID: 36006546 DOI: 10.1007/s11248-022-00325-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 08/09/2022] [Indexed: 01/20/2023]
Abstract
Glycogen storage disease type 1 (GSD1) is a rare hereditary monogenic disease characterized by the disturbed glucose metabolism. The most widespread variant of GSD1 is GSD1a, which is a deficiency of glucose-6-phosphatase-ɑ. Glucose-6-phosphatase-ɑ is expressed only in liver, kidney, and intestine, and these organs are primarily affected by its deficiency, and long-term complications of GSD1a include hepatic tumors and chronic liver disease. This article is a brief overview of existing animal models for GSD1a, from the first mouse model of 1996 to modern CRISPR/Cas9-generated ones. First whole-body murine models demonstrated exact metabolic symptoms of GSD1a, but the animals did not survive weaning. The protocol for glucose treatment allowed prolonged survival of affected animals, but long-term complications, such as hepatic tumorigenesis, could not be investigated. Next, organ-specific knockout models were developed, and most of the metabolic research was performed on liver glucose-6-phosphate-deficient mice. Naturally occuring mutation was also discovered in dogs. All these models are widely used to study GSD1a from metabolic and physiological standpoints and to develop possible treatments involving gene therapy. Research performed using these models helped elucidate the role of glycogen and lipid accumulation, hypoxia, mitochondrial dysfunction, and autophagy impairment in long-term complications of GSD1a, including hepatic tumorigenesis. Recently, gene replacement therapy and genome editing were tested on described models, and some of the developed approaches have reached clinical trials.
Collapse
Affiliation(s)
- Irina O Petrova
- Laboratory of Genome Editing, Research Center for Medical Genetics, Moskvorechye 1, Moscow, Russia, 115478.
| | - Svetlana A Smirnikhina
- Laboratory of Genome Editing, Research Center for Medical Genetics, Moskvorechye 1, Moscow, Russia, 115478
| |
Collapse
|
12
|
D’Acierno M, Resaz R, Iervolino A, Nielsen R, Sardella D, Siccardi S, Costanzo V, D’Apolito L, Suzumoto Y, Segalerba D, Astigiano S, Perna AF, Capasso G, Eva A, Trepiccione F. Dapagliflozin Prevents Kidney Glycogen Accumulation and Improves Renal Proximal Tubule Cell Functions in a Mouse Model of Glycogen Storage Disease Type 1b. J Am Soc Nephrol 2022; 33:1864-1875. [PMID: 35820785 PMCID: PMC9528317 DOI: 10.1681/asn.2021070935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 06/14/2022] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Mutations in SLC37A4, which encodes the intracellular glucose transporter G6PT, cause the rare glycogen storage disease type 1b (GSD1b). A long-term consequence of GSD1b is kidney failure, which requires KRT. The main protein markers of proximal tubule function, including NaPi2A, NHE3, SGLT2, GLUT2, and AQP1, are downregulated as part of the disease phenotype. METHODS We utilized an inducible mouse model of GSD1b, TM-G6PT-/-, to show that glycogen accumulation plays a crucial role in altering proximal tubule morphology and function. To limit glucose entry into proximal tubule cells and thus to prevent glycogen accumulation, we administered an SGLT2-inhibitor, dapagliflozin, to TM-G6PT-/- mice. RESULTS In proximal tubule cells, G6PT suppression stimulates the upregulation and activity of hexokinase-I, which increases availability of the reabsorbed glucose for intracellular metabolism. Dapagliflozin prevented glycogen accumulation and improved kidney morphology by promoting a metabolic switch from glycogen synthesis toward lysis and by restoring expression levels of the main proximal tubule functional markers. CONCLUSION We provide proof of concept for the efficacy of dapagliflozin in preserving kidney function in GSD1b mice. Our findings could represent the basis for repurposing this drug to treat patients with GSD1b.
Collapse
Affiliation(s)
| | - Roberta Resaz
- Laboratory of Molecular Biology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Anna Iervolino
- Biogem Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli,” Naples, Italy
| | - Rikke Nielsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Donato Sardella
- Biogem Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
| | - Sabrina Siccardi
- Biogem Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
| | - Vincenzo Costanzo
- Biogem Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
| | - Luciano D’Apolito
- Biogem Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
| | - Yoko Suzumoto
- Biogem Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
| | - Daniela Segalerba
- Laboratory of Molecular Biology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Alessandra F. Perna
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli,” Naples, Italy
| | - Giovambattista Capasso
- Biogem Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli,” Naples, Italy
| | - Alessandra Eva
- Laboratory of Molecular Biology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Francesco Trepiccione
- Biogem Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli,” Naples, Italy
| |
Collapse
|
13
|
Hotait ZS, Lo Cascio JN, Choos END, Shepard BD. The sugar daddy: the role of the renal proximal tubule in glucose homeostasis. Am J Physiol Cell Physiol 2022; 323:C791-C803. [PMID: 35912988 PMCID: PMC9448277 DOI: 10.1152/ajpcell.00225.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/26/2022] [Accepted: 07/26/2022] [Indexed: 11/22/2022]
Abstract
Renal blood flow represents >20% of total cardiac output and with this comes the great responsibility of maintaining homeostasis through the intricate regulation of solute handling. Through the processes of filtration, reabsorption, and secretion, the kidneys ensure that solutes and other small molecules are either returned to circulation, catabolized within renal epithelial cells, or excreted through the process of urination. Although this occurs throughout the renal nephron, one segment is tasked with the bulk of solute reabsorption-the proximal tubule. Among others, the renal proximal tubule is entirely responsible for the reabsorption of glucose, a critical source of energy that fuels the body. In addition, it is the only other site of gluconeogenesis outside of the liver. When these processes go awry, pathophysiological conditions such as diabetes and acidosis result. In this review, we highlight the recent advances made in understanding these processes that occur within the renal proximal tubule. We focus on the physiological mechanisms at play regarding glucose reabsorption and glucose metabolism, emphasize the conditions that occur under diseased states, and explore the emerging class of therapeutics that are responsible for restoring homeostasis.
Collapse
Affiliation(s)
- Zahraa S Hotait
- Department of Human Science, Georgetown University, Washington, District of Columbia
| | - Julia N Lo Cascio
- Department of Human Science, Georgetown University, Washington, District of Columbia
| | - Elijah N D Choos
- Department of Human Science, Georgetown University, Washington, District of Columbia
| | - Blythe D Shepard
- Department of Human Science, Georgetown University, Washington, District of Columbia
| |
Collapse
|
14
|
Mathis T, Poms M, Köfeler H, Gautschi M, Plecko B, Baumgartner MR, Hochuli M. Untargeted plasma metabolomics identifies broad metabolic perturbations in glycogen storage disease type I. J Inherit Metab Dis 2022; 45:235-247. [PMID: 34671989 PMCID: PMC9299190 DOI: 10.1002/jimd.12451] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND The metabolic defect in glycogen storage disease type I (GSDI) results in fasting hypoglycemia and typical secondary metabolic abnormalities (eg, hypertriglyceridemia, hyperlactatemia, hyperuricemia). The aim of this study was to assess further perturbations of the metabolic network in GSDI patients under ongoing treatment. METHODS In this prospective observational study, plasma samples of 14 adult patients (11 GSDIa, 3 GSDIb. Mean age 26.4 years, range 16-46 years) on standard treatment were compared to a cohort of 31 healthy controls utilizing ultra-high performance liquid chromatography (UHPLC) in combination with high resolution tandem mass spectrometry (HR-MS/MS) and subsequent statistical multivariate analysis. In addition, plasma fatty acid profiling was performed by GC/EI-MS. RESULTS The metabolomic profile showed alterations of metabolites in different areas of the metabolic network in both GSD subtypes, including pathways of fuel metabolism and energy generation, lipids and fatty acids, amino acid and methyl-group metabolism, the urea cycle, and purine/pyrimidine metabolism. These alterations were present despite adequate dietary treatment, did not correlate with plasma triglycerides or lactate, both parameters typically used to assess the quality of metabolic control in clinical practice, and were not related to the presence or absence of complications (ie, nephropathy or liver adenomas). CONCLUSION The metabolic defect of GSDI has profound effects on a variety of metabolic pathways in addition to the known typical abnormalities. These alterations are present despite optimized dietary treatment, which may contribute to the risk of developing long-term complications, an inherent problem of GSDI which appears to be only partly modified by current therapy.
Collapse
Affiliation(s)
- Tamara Mathis
- Division of Endocrinology, Diabetes, and Clinical NutritionUniversity Hospital Zurich, University of ZurichZurichSwitzerland
| | - Martin Poms
- Department of Clinical Chemistry and BiochemistryUniversity Children's Hospital Zurich, University of ZurichZurichSwitzerland
| | - Harald Köfeler
- Core Facility Mass SpectrometryMedical University of GrazGrazAustria
| | - Matthias Gautschi
- Division of Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics and Institute of Clinical ChemistryUniversity Hospital Bern, InselspitalBernSwitzerland
| | - Barbara Plecko
- Department of Pediatrics and Adolescent MedicineMedical University of GrazGrazAustria
| | - Matthias R. Baumgartner
- Division of Metabolism and Children's Research Center (CRC)University Children's Hospital, Zurich, University of ZurichZurichSwitzerland
- radiz—Rare Disease Initiative Zurich, Clinical Research Priority Program for Rare DiseasesUniversity of ZurichZurichSwitzerland
| | - Michel Hochuli
- Division of Endocrinology, Diabetes, and Clinical NutritionUniversity Hospital Zurich, University of ZurichZurichSwitzerland
- radiz—Rare Disease Initiative Zurich, Clinical Research Priority Program for Rare DiseasesUniversity of ZurichZurichSwitzerland
- Department of Diabetes, Endocrinology, Nutritional Medicine and MetabolismInselspital, Bern University Hospital and University of BernBernSwitzerland
| |
Collapse
|
15
|
Xu N, Han X, Zhang Y, Huang X, Zhu W, Shen M, Zhang W, Jialin C, Wei M, Qiu Z, Zeng X. Clinical features of gout in adult patients with type Ia glycogen storage disease: a single-centre retrospective study and a review of literature. Arthritis Res Ther 2022; 24:58. [PMID: 35219330 PMCID: PMC8881853 DOI: 10.1186/s13075-021-02706-5] [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] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/13/2021] [Indexed: 11/21/2022] Open
Abstract
Background This study aimed to explore the clinical features of gout in adult patients with glycogen storage disease type Ia (GSD Ia). Methods Ninety-five adult patients with GSD Ia admitted to Peking Union Medical College Hospital were retrospectively analysed. A clinical diagnosis of GSD Ia was confirmed in all patients through gene sequencing. All patients had hyperuricaemia; 31 patients complicated with gout were enrolled, and 64 adult GSD Ia patients with asymptomatic hyperuricaemia were selected as a control group during the same period. Clinical characteristics were analysed and compared between the two groups. Results Thirty-one of the 95 patients had complications of gout (median age, 25 years; 11 (35.5%) females). All 31 patients had hepatomegaly, abnormal liver function, fasting hypoglycaemia, hyperuricaemia, hyperlipaemia, and hyperlacticaemia. A protuberant abdomen, growth retardation, recurrent epistaxis, and diarrhoea were the most common clinical manifestations. Among these 31 patients, 10 patients (32.3%) had gout as the presenting manifestation and were diagnosed with GSD Ia at a median time of 5 years (range, 1–14) after the first gout flare. The median age of gout onset was 18 years (range, 10–29). Fifteen of the 31 GSD Ia-related gout patients were complicated with gouty tophi, which has an average incidence time of 2 years after the first gouty flare. The mean value of the maximum serum uric acid (SUA) was 800.5 μmol/L (range, 468–1068). The incidence of gout in adult GSD Ia patients was significantly associated with the initial age of regular treatment with raw corn starch, the proportion of urate-lowering therapy initiated during the asymptomatic hyperuricaemic stage, maximum SUA level, and mean cholesterol level. Conclusions Determination of GSD Ia should be performed for young-onset gout patients with an early occurrence of gouty tophi, especially in patients with hepatomegaly, recurrent hypoglycaemia, or growth retardation. Early detection and long-term regulatory management of hyperuricaemia, in addition to early raw corn starch and lifestyle intervention, should be emphasized for GSD Ia patients in order to maintain good metabolic control. Trial registration Retrospectively registered.
Collapse
Affiliation(s)
- Na Xu
- Department of family medicine & Division of General Internal Medicine, Department of medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases (Peking Union Medical College Hospital), Beijing, China
| | - Xinxin Han
- Department of family medicine & Division of General Internal Medicine, Department of medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases (Peking Union Medical College Hospital), Beijing, China
| | - Yun Zhang
- Department of family medicine & Division of General Internal Medicine, Department of medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases (Peking Union Medical College Hospital), Beijing, China
| | - Xiaoming Huang
- Department of family medicine & Division of General Internal Medicine, Department of medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases (Peking Union Medical College Hospital), Beijing, China
| | - Weiguo Zhu
- Department of family medicine & Division of General Internal Medicine, Department of medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases (Peking Union Medical College Hospital), Beijing, China
| | - Min Shen
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Wen Zhang
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Chen Jialin
- Department of family medicine & Division of General Internal Medicine, Department of medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases (Peking Union Medical College Hospital), Beijing, China
| | - Min Wei
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhengqing Qiu
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Xuejun Zeng
- Department of family medicine & Division of General Internal Medicine, Department of medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases (Peking Union Medical College Hospital), Beijing, China.
| |
Collapse
|
16
|
La Rose AM, Bazioti V, Hoogerland JA, Svendsen AF, Groenen AG, van Faassen M, Rutten MGS, Kloosterhuis NJ, Dethmers-Ausema B, Nijland JH, Mithieux G, Rajas F, Kuipers F, Lukens MV, Soehnlein O, Oosterveer MH, Westerterp M. Hepatocyte-specific glucose-6-phosphatase deficiency disturbs platelet aggregation and decreases blood monocytes upon fasting-induced hypoglycemia. Mol Metab 2021; 53:101265. [PMID: 34091064 PMCID: PMC8243524 DOI: 10.1016/j.molmet.2021.101265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/20/2021] [Accepted: 05/31/2021] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Glycogen storage disease type 1a (GSD Ia) is a rare inherited metabolic disorder caused by mutations in the glucose-6-phosphatase (G6PC1) gene. When untreated, GSD Ia leads to severe fasting-induced hypoglycemia. Although current intensive dietary management aims to prevent hypoglycemia, patients still experience hypoglycemic events. Poor glycemic control in GSD Ia is associated with hypertriglyceridemia, hepatocellular adenoma and carcinoma, and also with an increased bleeding tendency of unknown origin. METHODS To evaluate the effect of glycemic control on leukocyte levels and coagulation in GSD Ia, we employed hepatocyte-specific G6pc1 deficient (L-G6pc-/-) mice under fed or fasted conditions, to match good or poor glycemic control in GSD Ia, respectively. RESULTS We found that fasting-induced hypoglycemia in L-G6pc-/- mice decreased blood leukocytes, specifically proinflammatory Ly6Chi monocytes, compared to controls. Refeeding reversed this decrease. The decrease in Ly6Chi monocytes was accompanied by an increase in plasma corticosterone levels and was prevented by the glucocorticoid receptor antagonist mifepristone. Further, fasting-induced hypoglycemia in L-G6pc-/- mice prolonged bleeding time in the tail vein bleeding assay, with reversal by refeeding. This could not be explained by changes in coagulation factors V, VII, or VIII, or von Willebrand factor. While the prothrombin and activated partial thromboplastin time as well as total platelet counts were not affected by fasting-induced hypoglycemia in L-G6pc-/- mice, ADP-induced platelet aggregation was disturbed. CONCLUSIONS These studies reveal a relationship between fasting-induced hypoglycemia, decreased blood monocytes, and disturbed platelet aggregation in L-G6pc-/- mice. While disturbed platelet aggregation likely accounts for the bleeding phenotype in GSD Ia, elevated plasma corticosterone decreases the levels of proinflammatory monocytes. These studies highlight the necessity of maintaining good glycemic control in GSD Ia.
Collapse
Affiliation(s)
- Anouk M La Rose
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Venetia Bazioti
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Joanne A Hoogerland
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Arthur F Svendsen
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anouk G Groenen
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Martijn van Faassen
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Martijn G S Rutten
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Niels J Kloosterhuis
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Bertien Dethmers-Ausema
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - J Hendrik Nijland
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Gilles Mithieux
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Fabienne Rajas
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Folkert Kuipers
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Michaël V Lukens
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Oliver Soehnlein
- Institute for Experimental Pathology (ExPat), Center for Molecular Biology of Inflammation (ZBME), University of Münster, Münster, Germany; Department of Physiology and Pharmacology (FyFa), Karolinska Institutet, Stockholm, Sweden
| | - Maaike H Oosterveer
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marit Westerterp
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
17
|
Monteillet L, Labrune P, Hochuli M, Do Cao J, Tortereau A, Miliano AC, Ardon-Zitoun C, Duchampt A, Silva M, Verzieux V, Mithieux G, Rajas F. Cellular and metabolic effects of renin-angiotensin system blockade on glycogen storage disease type I nephropathy. Hum Mol Genet 2021; 31:914-928. [PMID: 34617103 PMCID: PMC8947214 DOI: 10.1093/hmg/ddab297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/04/2021] [Accepted: 10/04/2021] [Indexed: 02/06/2023] Open
Abstract
Glycogen Storage Disease Type I (GSDI) is an inherited disease caused by glucose-6 phosphatase (G6Pase) deficiency, leading to a loss of endogenous glucose production and severe hypoglycemia. Moreover, most GSDI patients develop a chronic kidney disease (CKD) due to lipid accumulation in the kidney. Similar to diabetic CKD, activation of renin-angiotensin system (RAS) promotes renal fibrosis in GSDI. Here, we investigated the physiological and molecular effects of RAS blockers in GSDI patients and mice. A retrospective analysis of renal function was performed in 21 GSDI patients treated with RAS blockers. Cellular and metabolic impacts of RAS blockade were analyzed in K.G6pc−/− mice characterized by G6pc1 deletion in kidneys. GSDI patients started RAS blocker treatment at a median age of 21 years and long-term treatment reduced the progression of CKD in about 50% of patients. However, CKD progressed to kidney failure in 20% of treated patients, requiring renal transplantation. In K.G6pc−/− mice, CKD was associated with an impairment of autophagy and ER stress. RAS blockade resulted in a rescue of autophagy and decreased ER stress, concomitantly with decreased fibrosis and improved renal function, but without impact on glycogen and lipid contents. In conclusion, these data confirm the partial beneficial effect of RAS blockers in the prevention of CKD in GSDI. Mechanistically, we show that these effects are linked to a reduction of cell stress, without affecting metabolism.
Collapse
Affiliation(s)
- Laure Monteillet
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Philippe Labrune
- APHP, Université Paris-Saclay, Hôpital Antoine Béclère, Clamart, France
| | - Michel Hochuli
- Department of Diabetes, Endocrinology, Nutritional Medicine and Metabolism, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Jeremy Do Cao
- APHP, Université Paris-Saclay, Hôpital Antoine Béclère, Clamart, France
| | | | | | - Carine Ardon-Zitoun
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Adeline Duchampt
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Marine Silva
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Vincent Verzieux
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Gilles Mithieux
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Fabienne Rajas
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| |
Collapse
|
18
|
Osada Y, Nakagawa S, Ishibe K, Takao S, Shimazaki A, Itohara K, Imai S, Yonezawa A, Nakagawa T, Matsubara K. Antibiotic-induced microbiome depletion alters renal glucose metabolism and exacerbates renal injury after ischemia-reperfusion injury in mice. Am J Physiol Renal Physiol 2021; 321:F455-F465. [PMID: 34423680 DOI: 10.1152/ajprenal.00111.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Recent studies have revealed the impact of antibiotic-induced microbiome depletion (AIMD) on host glucose homeostasis. The kidney has a critical role in systemic glucose homeostasis; however, information regarding the association between AIMD and renal glucose metabolism remains limited. Hence, we aimed to determine the effects of AIMD on renal glucose metabolism by inducing gut microbiome depletion using an antibiotic cocktail (ABX) composed of ampicillin, vancomycin, and levofloxacin in mice. The results showed that bacterial 16s rRNA expression, luminal concentrations of short-chain fatty acids and bile acids, and plasma glucose levels were significantly lower in ABX-treated mice than in vehicle-treated mice. In addition, ABX treatment significantly reduced renal glucose and pyruvate levels. mRNA expression levels of glucose-6-phosphatase and phosphoenolpyruvate carboxykinase in the renal cortex were significantly higher in ABX-treated mice than in vehicle-treated mice. We further examined the impact of AIMD on the altered metabolic status in mice after ischemia-induced kidney injury. After exposure to ischemia for 60 min, renal pyruvate concentrations were significantly lower in ABX-treated mice than in vehicle-treated mice. ABX treatment caused a more severe tubular injury after ischemia-reperfusion. Our findings confirm that AIMD is associated with decreased pyruvate levels in the kidney, which may have been caused by the activation of renal gluconeogenesis. Thus, we hypothesized that AIMD would increase the vulnerability of the kidney to ischemia-reperfusion injury.NEW & NOTEWORTHY This study aimed to determine the impact of antibiotic-induced microbiome depletion (AIMD) on renal glucose metabolism in mice. This is the first report confirming that AIMD is associated with decreased levels of pyruvate, a key intermediate in glucose metabolism, which may have been caused by activation of renal gluconeogenesis. We hypothesized that AIMD can increase the susceptibility of the kidney to ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Yuika Osada
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Shunsaku Nakagawa
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Kanako Ishibe
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Shota Takao
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Aimi Shimazaki
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Kotaro Itohara
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Satoshi Imai
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Atsushi Yonezawa
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Takayuki Nakagawa
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Kazuo Matsubara
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
19
|
Jia S, Peng X, Liang L, Zhang Y, Li M, Zhou Q, Shen X, Wang Y, Wang C, Feng S, Chen J, Ren P, Jiang H. The Study of Angptl4-Modulated Podocyte Injury in IgA Nephropathy. Front Physiol 2021; 11:575722. [PMID: 33643055 PMCID: PMC7905042 DOI: 10.3389/fphys.2020.575722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/20/2020] [Indexed: 11/16/2022] Open
Abstract
Background Increasing evidence shows that Angptl4 affects proteinuria in podocytes injured kidney disease, however, whether there is a relationship between Angptl4 and IgA nephropathy (IgAN) has not been studied yet. Methods Plasma and urine samples were obtained from 71 patients with IgAN and 61 healthy controls. Glomeruli from six renal biopsy specimens (three IgAN patients and three healthy controls) were separated by RNA-Seq. Differentially expressed genes (DEGs) related to podocytes and Angptl4 between IgAN patients and healthy controls were performed using the Limma package. Gene set enrichment analysis was used to determine whether there was a statistically significant difference between the two groups. STRING was used to create a protein-protein interaction network of DEGs. Association analysis between Angptl4 levels and clinical features of IgAN was performed. Results Thirty-three podocyte-related and twenty-three Angpt4-related DEGs were found between IgAN patients and healthy controls. By overlapping the genes, FOS and G6PC were found to be upregulated in IgAN patients, while MMP9 was downregulated in IgAN patients. Plasma and urine Angptl4 levels were closely related to the degree of podocyte injury and urine protein, but not to the protein-creatine ratio. Conclusion Our findings show that Angptl4 levels in plasma and urine are related to podocyte damage and, therefore, may be a promising tool for assessing the severity of IgAN patients to identify and reverse the progression to ESRD.
Collapse
Affiliation(s)
- Sha Jia
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Nephropathy, Hangzhou, China.,Kidney Disease Immunology Laboratory, The Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Beijing, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China.,Dongyang Women & Children Hospital, Dongyang, China
| | - Xiaofeng Peng
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Nephropathy, Hangzhou, China.,Kidney Disease Immunology Laboratory, The Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Beijing, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Ludan Liang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Nephropathy, Hangzhou, China.,Kidney Disease Immunology Laboratory, The Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Beijing, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Ying Zhang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Nephropathy, Hangzhou, China.,Kidney Disease Immunology Laboratory, The Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Beijing, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Meng Li
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Nephropathy, Hangzhou, China.,Kidney Disease Immunology Laboratory, The Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Beijing, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Qin Zhou
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Nephropathy, Hangzhou, China.,Kidney Disease Immunology Laboratory, The Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Beijing, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Xiujin Shen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Nephropathy, Hangzhou, China.,Kidney Disease Immunology Laboratory, The Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Beijing, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Yucheng Wang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Nephropathy, Hangzhou, China.,Kidney Disease Immunology Laboratory, The Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Beijing, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Cuili Wang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Nephropathy, Hangzhou, China.,Kidney Disease Immunology Laboratory, The Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Beijing, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Shi Feng
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Nephropathy, Hangzhou, China.,Kidney Disease Immunology Laboratory, The Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Beijing, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Nephropathy, Hangzhou, China.,Kidney Disease Immunology Laboratory, The Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Beijing, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Pingping Ren
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Nephropathy, Hangzhou, China.,Kidney Disease Immunology Laboratory, The Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Beijing, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Hong Jiang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Nephropathy, Hangzhou, China.,Kidney Disease Immunology Laboratory, The Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Beijing, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China
| |
Collapse
|
20
|
Lee YH, Seo JW, Kim M, Tae D, Seok J, Kim YG, Lee SH, Kim JS, Hwang HS, Jeong KH, Moon JY. Urinary mRNA Signatures as Predictors of Renal Function Decline in Patients With Biopsy-Proven Diabetic Kidney Disease. Front Endocrinol (Lausanne) 2021; 12:774436. [PMID: 34858345 PMCID: PMC8630698 DOI: 10.3389/fendo.2021.774436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 10/12/2021] [Indexed: 01/12/2023] Open
Abstract
The clinical manifestations of diabetic kidney disease (DKD) are more heterogeneous than those previously reported, and these observations mandate the need for the recruitment of patients with biopsy-proven DKD in biomarker research. In this study, using the public gene expression omnibus (GEO) repository, we aimed to identify urinary mRNA biomarkers that can predict histological severity and disease progression in patients with DKD in whom the diagnosis and histologic grade has been confirmed by kidney biopsy. We identified 30 DKD-specific mRNA candidates based on the analysis of the GEO datasets. Among these, there were significant alterations in the urinary levels of 17 mRNAs in patients with DKD, compared with healthy controls. Four urinary mRNAs-LYZ, C3, FKBP5, and G6PC-reflected tubulointerstitial inflammation and fibrosis in kidney biopsy and could predict rapid progression to end-stage kidney disease independently of the baseline eGFR (tertile 1 vs. tertile 3; adjusted hazard ratio of 9.68 and 95% confidence interval of 2.85-32.87, p < 0.001). In conclusion, we demonstrated that urinary mRNA signatures have a potential to indicate the pathologic status and predict adverse renal outcomes in patients with DKD.
Collapse
Affiliation(s)
- Yu Ho Lee
- Division of Nephrology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Jung-Woo Seo
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University School of Medicine, Seoul, South Korea
| | - Miji Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University School of Medicine, Seoul, South Korea
| | - Donghyun Tae
- School of Electrical Engineering, Korea University, Seoul, South Korea
| | - Junhee Seok
- School of Electrical Engineering, Korea University, Seoul, South Korea
| | - Yang Gyun Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University School of Medicine, Seoul, South Korea
| | - Sang-Ho Lee
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University School of Medicine, Seoul, South Korea
| | - Jin Sug Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University School of Medicine, Seoul, South Korea
| | - Hyeon Seok Hwang
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University School of Medicine, Seoul, South Korea
| | - Kyung-Hwan Jeong
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University School of Medicine, Seoul, South Korea
| | - Ju-Young Moon
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University School of Medicine, Seoul, South Korea
- *Correspondence: Ju-Young Moon,
| |
Collapse
|
21
|
Almodóvar-Payá A, Villarreal-Salazar M, de Luna N, Nogales-Gadea G, Real-Martínez A, Andreu AL, Martín MA, Arenas J, Lucia A, Vissing J, Krag T, Pinós T. Preclinical Research in Glycogen Storage Diseases: A Comprehensive Review of Current Animal Models. Int J Mol Sci 2020; 21:ijms21249621. [PMID: 33348688 PMCID: PMC7766110 DOI: 10.3390/ijms21249621] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 12/19/2022] Open
Abstract
GSD are a group of disorders characterized by a defect in gene expression of specific enzymes involved in glycogen breakdown or synthesis, commonly resulting in the accumulation of glycogen in various tissues (primarily the liver and skeletal muscle). Several different GSD animal models have been found to naturally present spontaneous mutations and others have been developed and characterized in order to further understand the physiopathology of these diseases and as a useful tool to evaluate potential therapeutic strategies. In the present work we have reviewed a total of 42 different animal models of GSD, including 26 genetically modified mouse models, 15 naturally occurring models (encompassing quails, cats, dogs, sheep, cattle and horses), and one genetically modified zebrafish model. To our knowledge, this is the most complete list of GSD animal models ever reviewed. Importantly, when all these animal models are analyzed together, we can observe some common traits, as well as model specific differences, that would be overlooked if each model was only studied in the context of a given GSD.
Collapse
Affiliation(s)
- Aitana Almodóvar-Payá
- Mitochondrial and Neuromuscular Disorders Unit, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (A.A.-P.); (M.V.-S.); (A.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; (N.d.L.); (G.N.-G.); (M.A.M.); (J.A.)
| | - Mónica Villarreal-Salazar
- Mitochondrial and Neuromuscular Disorders Unit, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (A.A.-P.); (M.V.-S.); (A.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; (N.d.L.); (G.N.-G.); (M.A.M.); (J.A.)
| | - Noemí de Luna
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; (N.d.L.); (G.N.-G.); (M.A.M.); (J.A.)
- Laboratori de Malalties Neuromusculars, Institut de Recerca Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, 08041 Barcelona, Spain
| | - Gisela Nogales-Gadea
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; (N.d.L.); (G.N.-G.); (M.A.M.); (J.A.)
- Grup de Recerca en Malalties Neuromusculars i Neuropediàtriques, Department of Neurosciences, Institut d’Investigacio en Ciencies de la Salut Germans Trias i Pujol i Campus Can Ruti, Universitat Autònoma de Barcelona, 08916 Badalona, Spain
| | - Alberto Real-Martínez
- Mitochondrial and Neuromuscular Disorders Unit, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (A.A.-P.); (M.V.-S.); (A.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; (N.d.L.); (G.N.-G.); (M.A.M.); (J.A.)
| | - Antoni L. Andreu
- EATRIS, European Infrastructure for Translational Medicine, 1081 HZ Amsterdam, The Netherlands;
| | - Miguel Angel Martín
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; (N.d.L.); (G.N.-G.); (M.A.M.); (J.A.)
- Mitochondrial and Neuromuscular Diseases Laboratory, 12 de Octubre Hospital Research Institute (i+12), 28041 Madrid, Spain
| | - Joaquin Arenas
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; (N.d.L.); (G.N.-G.); (M.A.M.); (J.A.)
- Mitochondrial and Neuromuscular Diseases Laboratory, 12 de Octubre Hospital Research Institute (i+12), 28041 Madrid, Spain
| | - Alejandro Lucia
- Faculty of Sport Sciences, European University, 28670 Madrid, Spain;
| | - John Vissing
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark; (J.V.); (T.K.)
| | - Thomas Krag
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark; (J.V.); (T.K.)
| | - Tomàs Pinós
- Mitochondrial and Neuromuscular Disorders Unit, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (A.A.-P.); (M.V.-S.); (A.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; (N.d.L.); (G.N.-G.); (M.A.M.); (J.A.)
- Correspondence: ; Tel.: +34-934894057
| |
Collapse
|
22
|
Rasal KD, Iquebal MA, Dixit S, Vasam M, Raza M, Sahoo L, Jaiswal S, Nandi S, Mahapatra KD, Rasal A, Udit UK, Meher PK, Murmu K, Angadi UB, Rai A, Kumar D, Sundaray JK. Revealing Alteration in the Hepatic Glucose Metabolism of Genetically Improved Carp, Jayanti Rohu Labeo rohita Fed a High Carbohydrate Diet Using Transcriptome Sequencing. Int J Mol Sci 2020; 21:E8180. [PMID: 33142948 PMCID: PMC7662834 DOI: 10.3390/ijms21218180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 01/25/2023] Open
Abstract
Although feed cost is the greatest concern in aquaculture, the inclusion of carbohydrates in the fish diet, and their assimilation, are still not well understood in aquaculture species. We identified molecular events that occur due to the inclusion of high carbohydrate levels in the diets of genetically improved 'Jayanti rohu' Labeo rohita. To reveal transcriptional changes in the liver of rohu, a feeding experiment was conducted with three doses of gelatinized starch (20% (control), 40%, and 60%). Transcriptome sequencing revealed totals of 15,232 (4464 up- and 4343 down-regulated) and 15,360 (4478 up- and 4171 down-regulated) differentially expressed genes. Up-regulated transcripts associated with glucose metabolisms, such as hexokinase, PHK, glycogen synthase and PGK, were found in fish fed diets with high starch levels. Interestingly, a de novo lipogenesis mechanism was found to be enriched in the livers of treated fish due to up-regulated transcripts such as FAS, ACCα, and PPARγ. The insulin signaling pathways with enriched PPAR and mTOR were identified by Kyoto Encyclopedia of Genes and Genome (KEGG) as a result of high carbohydrates. This work revealed for the first time the atypical regulation transcripts associated with glucose metabolism and lipogenesis in the livers of Jayanti rohu due to the inclusion of high carbohydrate levels in the diet. This study also encourages the exploration of early nutritional programming for enhancing glucose efficiency in carp species, for sustainable and cost-effective aquaculture production.
Collapse
Affiliation(s)
- Kiran D. Rasal
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Mir Asif Iquebal
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Sangita Dixit
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Manohar Vasam
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Mustafa Raza
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Lakshman Sahoo
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Sarika Jaiswal
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Samiran Nandi
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Kanta Das Mahapatra
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Avinash Rasal
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Uday Kumar Udit
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Prem Kumar Meher
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Khuntia Murmu
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - UB Angadi
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Anil Rai
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Dinesh Kumar
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Jitendra Kumar Sundaray
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| |
Collapse
|
23
|
Perry A, Douillard C, Jonca F, Glowacki F, Leroy X, Caveriviere P, Hubert A, Labrune P. Papillary renal cell carcinoma in two young adults with glycogen storage disease type Ia. JIMD Rep 2020; 52:17-22. [PMID: 32154055 PMCID: PMC7052693 DOI: 10.1002/jmd2.12096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 01/06/2020] [Accepted: 01/10/2020] [Indexed: 01/30/2023] Open
Abstract
Glycogen storage disease type Ia (GSD Ia) is a rare metabolic disease due to glucose‐6‐phosphatase deficiency. Chronic kidney disease is a frequent complication that may manifest itself by glomerular lesions and tubular dysfunction from the second decade of life. We report two young GSDIa patients with malignant renal tumor. The first patient was a 25‐year‐old man. He had chronic metabolic imbalance without kidney involvement. The tumor, a type 2 papillary renal carcinoma, was accidentally discovered during follow‐up. The second patient was a 27‐year‐old woman with chronic metabolic imbalance and chronic kidney involvement. The tumor, a grade 2 papillary carcinoma, was accidentally discovered during follow‐up. These two observations are, to date, the first to be reported. We suggest that annual monitoring of kidney imaging in GSDI patients should be systematic to detect renal cancer, from the second decade of life.
Collapse
Affiliation(s)
- Ariane Perry
- APHP, Hôpitaux Universitaires Paris Sud, Hôpital Antoine Béclère, Centre de référence des maladies héréditaires du métabolisme hépatique Clamart France
| | - Claire Douillard
- Lille University Hospital, Hôpital Jeanne de Flandres, Centre de référence des maladies héréditaires du métabolisme Lille France
| | | | - Francois Glowacki
- Nephrology Department Huriez Hospital, Lille University Hospital Lille France
| | - Xavier Leroy
- Department of Pathology Univ. Lille, CHU Lille France
| | - Paul Caveriviere
- Anatomy and pathology laboratory, les Feuillants Toulouse France
| | - Aurélie Hubert
- APHP, Hôpitaux Universitaires Paris Sud, Hôpital Antoine Béclère, Centre de référence des maladies héréditaires du métabolisme hépatique Clamart France
| | - Philippe Labrune
- APHP, Hôpitaux Universitaires Paris Sud, Hôpital Antoine Béclère, Centre de référence des maladies héréditaires du métabolisme hépatique Clamart France.,Université Paris Sud Paris Saclay and INSERM U 1195 France
| |
Collapse
|
24
|
Jauze L, Monteillet L, Mithieux G, Rajas F, Ronzitti G. Challenges of Gene Therapy for the Treatment of Glycogen Storage Diseases Type I and Type III. Hum Gene Ther 2019; 30:1263-1273. [PMID: 31319709 DOI: 10.1089/hum.2019.102] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Glycogen storage diseases (GSDs) type I (GSDI) and type III (GSDIII), the most frequent hepatic GSDs, are due to defects in glycogen metabolism, mainly in the liver. In addition to hypoglycemia and liver pathology, renal, myeloid, or muscle complications affect GSDI and GSDIII patients. Currently, patient management is based on dietary treatment preventing severe hypoglycemia and increasing the lifespan of patients. However, most of the patients develop long-term pathologies. In the past years, gene therapy for GSDI has generated proof of concept for hepatic GSDs. This resulted in a recent clinical trial of adeno-associated virus (AAV)-based gene replacement for GSDIa. However, the current limitations of AAV-mediated gene transfer still represent a challenge for successful gene therapy in GSDI and GSDIII. Indeed, transgene loss over time was observed in GSDI liver, possibly due to the degeneration of hepatocytes underlying the physiopathology of both GSDI and GSDIII and leading to hepatic tumor development. Moreover, multitissue targeting requires high vector doses to target nonpermissive tissues such as muscle and kidney. Interestingly, recent pharmacological interventions or dietary regimen aiming at the amelioration of the hepatocyte abnormalities before the administration of gene therapy demonstrated improved efficacy in GSDs. In this review, we describe the advances in gene therapy and the limitations to be overcome to achieve efficient and safe gene transfer in GSDs.
Collapse
Affiliation(s)
- Louisa Jauze
- INTEGRARE, Genethon, Inserm, Université d'Evry, Université Paris-Saclay, Evry, France.,Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, France.,Université de Lyon, Lyon, France.,Université Lyon I, Villeurbanne, France
| | - Laure Monteillet
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, France.,Université de Lyon, Lyon, France.,Université Lyon I, Villeurbanne, France
| | - Gilles Mithieux
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, France.,Université de Lyon, Lyon, France.,Université Lyon I, Villeurbanne, France
| | - Fabienne Rajas
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, France.,Université de Lyon, Lyon, France.,Université Lyon I, Villeurbanne, France
| | - Giuseppe Ronzitti
- INTEGRARE, Genethon, Inserm, Université d'Evry, Université Paris-Saclay, Evry, France
| |
Collapse
|
25
|
Brooks ED, Landau DJ, Everitt JI, Brown TT, Grady KM, Waskowicz L, Bass CR, D'Angelo J, Asfaw YG, Williams K, Kishnani PS, Koeberl DD. Long-term complications of glycogen storage disease type Ia in the canine model treated with gene replacement therapy. J Inherit Metab Dis 2018; 41:965-976. [PMID: 30043186 PMCID: PMC6328337 DOI: 10.1007/s10545-018-0223-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 06/09/2018] [Accepted: 06/19/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND Glycogen storage disease type Ia (GSD Ia) in dogs closely resembles human GSD Ia. Untreated patients with GSD Ia develop complications associated with glucose-6-phosphatase (G6Pase) deficiency. Survival of human patients on intensive nutritional management has improved; however, long-term complications persist including renal failure, nephrolithiasis, hepatocellular adenomas (HCA), and a high risk for hepatocellular carcinoma (HCC). Affected dogs fail to thrive with dietary therapy alone. Treatment with gene replacement therapy using adeno-associated viral vectors (AAV) expressing G6Pase has greatly prolonged life and prevented hypoglycemia in affected dogs. However, long-term complications have not been described to date. METHODS Five GSD Ia-affected dogs treated with AAV-G6Pase were evaluated. Dogs were euthanized due to reaching humane endpoints related to liver and/or kidney involvement, at 4 to 8 years of life. Necropsies were performed and tissues were analyzed. RESULTS Four dogs had liver tumors consistent with HCA and HCC. Three dogs developed renal failure, but all dogs exhibited progressive kidney disease histologically. Urolithiasis was detected in two dogs; uroliths were composed of calcium oxalate and calcium phosphate. One affected and one carrier dog had polycystic ovarian disease. Bone mineral density was not significantly affected. CONCLUSIONS Here, we show that the canine GSD Ia model demonstrates similar long-term complications as GSD Ia patients in spite of gene replacement therapy. Further development of gene therapy is needed to develop a more effective treatment to prevent long-term complications of GSD Ia.
Collapse
Affiliation(s)
- Elizabeth D Brooks
- Division of Medical Genetics, Duke University Medical Center (DUMC), Box 103856, Durham, NC, 27710, USA
- Division of Laboratory Animal Resources, Duke University Medical Center, Durham, NC, USA
| | - Dustin J Landau
- Division of Medical Genetics, Duke University Medical Center (DUMC), Box 103856, Durham, NC, 27710, USA
| | - Jeffrey I Everitt
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Talmage T Brown
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Kylie M Grady
- Division of Medical Genetics, Duke University Medical Center (DUMC), Box 103856, Durham, NC, 27710, USA
| | - Lauren Waskowicz
- Division of Medical Genetics, Duke University Medical Center (DUMC), Box 103856, Durham, NC, 27710, USA
| | - Cameron R Bass
- Department of Biomedical Engineering, Duke University Medical Center, Durham, NC, USA
| | - John D'Angelo
- Department of Biomedical Engineering, Duke University Medical Center, Durham, NC, USA
| | - Yohannes G Asfaw
- Division of Laboratory Animal Resources, Duke University Medical Center, Durham, NC, USA
| | - Kyha Williams
- Division of Laboratory Animal Resources, Duke University Medical Center, Durham, NC, USA
| | - Priya S Kishnani
- Division of Medical Genetics, Duke University Medical Center (DUMC), Box 103856, Durham, NC, 27710, USA
| | - Dwight D Koeberl
- Division of Medical Genetics, Duke University Medical Center (DUMC), Box 103856, Durham, NC, 27710, USA.
| |
Collapse
|
26
|
Gjorgjieva M, Monteillet L, Calderaro J, Mithieux G, Rajas F. Polycystic kidney features of the renal pathology in glycogen storage disease type I: possible evolution to renal neoplasia. J Inherit Metab Dis 2018; 41:955-963. [PMID: 29869165 DOI: 10.1007/s10545-018-0207-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/07/2018] [Accepted: 05/22/2018] [Indexed: 12/12/2022]
Abstract
Glycogen storage disease type I (GSDI) is a rare genetic pathology characterized by glucose-6 phosphatase (G6Pase) deficiency, translating in hypoglycemia during short fasts. Besides metabolic perturbations, GSDI patients develop long-term complications, especially chronic kidney disease (CKD). In GSDI patients, CKD is characterized by an accumulation of glycogen and lipids in kidneys, leading to a gradual decline in renal function. At a molecular level, the activation of the renin-angiotensin system is responsible for the development of renal fibrosis, eventually leading to renal failure. The same CKD phenotype was observed in a mouse model with a kidney-specific G6Pase deficiency (K.G6pc-/- mice). Furthermore, GSDI patients and mice develop frequently renal cysts at late stages of the nephropathy, classifying GSDI as a potential polycystic kidney disease (PKD). PKDs are genetic disorders characterized by multiple renal cyst formation, frequently caused by the loss of expression of polycystic kidney genes, such as PKD1/2 and PKHD1. Interestingly, these genes are deregulated in K.G6pc-/- kidneys, suggesting their possible role in GSDI cystogenesis. Finally, renal cysts are known to predispose to renal malignancy development. In addition, HNF1B loss is a malignancy prediction factor. Interestingly, Hnf1b expression was decreased in K.G6pc-/- kidneys. While a single case of renal cancer has been reported in a GSDI patient, a clear cell renal carcinoma was recently observed in one K.G6pc-/- mouse (out of 36 studied mice) at a later stage of the disease. This finding highlights the need to further analyze renal cyst development in GSDI patients in order to evaluate the possible associated risk of carcinogenesis, even if the risk might be limited.
Collapse
Affiliation(s)
- Monika Gjorgjieva
- Institut National de la Santé et de la Recherche by Inserm, U1213, 69008, Lyon, France
- Université de Lyon, 69008, Lyon, France
- Université Lyon1, 69622, Villeurbanne, France
| | - Laure Monteillet
- Institut National de la Santé et de la Recherche by Inserm, U1213, 69008, Lyon, France
- Université de Lyon, 69008, Lyon, France
- Université Lyon1, 69622, Villeurbanne, France
| | - Julien Calderaro
- Inserm UMR-1162, Université Paris Descartes, Labex Immuno-Oncology, Université Paris Diderot, Université Paris 13, Paris, France
- APHP, Assistance-Publique Hôpitaux-de-Paris, Département de Pathologie, Hôpital Henri Mondor, 94010, Créteil, France
| | - Gilles Mithieux
- Institut National de la Santé et de la Recherche by Inserm, U1213, 69008, Lyon, France
- Université de Lyon, 69008, Lyon, France
- Université Lyon1, 69622, Villeurbanne, France
| | - Fabienne Rajas
- Institut National de la Santé et de la Recherche by Inserm, U1213, 69008, Lyon, France.
- Université de Lyon, 69008, Lyon, France.
- Université Lyon1, 69622, Villeurbanne, France.
- Inserm U1213, Université Lyon 1 Laennec, 7 rue Guillaume Paradin, 69372, Lyon Cedex 08, France.
| |
Collapse
|
27
|
Lee YM, Conlon TJ, Specht A, Coleman KE, Brown LM, Estrella AM, Dambska M, Dahlberg KR, Weinstein DA. Long-term safety and efficacy of AAV gene therapy in the canine model of glycogen storage disease type Ia. J Inherit Metab Dis 2018; 41:977-984. [PMID: 29802554 DOI: 10.1007/s10545-018-0199-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 04/30/2018] [Accepted: 05/14/2018] [Indexed: 01/25/2023]
Abstract
BACKGROUND Viral mediated gene therapy has progressed after overcoming early failures, and gene therapy has now been approved for several conditions in Europe and the USA. Glycogen storage disease (GSD) type Ia, caused by a deficiency of glucose-6-phosphatase-α, has been viewed as an outstanding candidate for gene therapy. This follow-up report describes the long-term outcome for the naturally occurring GSD-Ia dogs treated with rAAV-GPE-hG6PC-mediated gene therapy. METHODS A total of seven dogs were treated with rAAV-GPE-hG6PC-mediated gene therapy. The first four dogs were treated at birth, and three dogs were treated between 2 and 6 months of age to assess the efficacy and safety in animals with mature livers. Blood and urine samples, radiographic studies, histological evaluation, and biodistribution were assessed. RESULTS Gene therapy improved survival in the GSD-Ia dogs. With treatment, the biochemical studies normalized for the duration of the study (up to 7 years). None of the rAAV-GPE-hG6PC-treated dogs had focal hepatic lesions or renal abnormalities. Dogs treated at birth required a second dose of rAAV after 2-4 months; gene therapy after hepatic maturation resulted in improved efficacy after a single dose. CONCLUSION rAAV-GPE-hG6PC treatment in GSD-Ia dogs was found to be safe and efficacious. GSD-Ia is an attractive target for human gene therapy since it is a monogenic disorder with limited tissue involvement. Blood glucose and lactate monitoring can be used to assess effectiveness and as a biomarker of success. GSD-Ia can also serve as a model for other hepatic monogenic disorders.
Collapse
Affiliation(s)
- Young Mok Lee
- Glycogen Storage Disease Program, Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
- Glycogen Storage Disease Program, University of Florida College of Medicine, Gainesville, FL, USA
| | - Thomas J Conlon
- Powell Gene Therapy Center, University of Florida, Gainesville, FL, 32610, USA
- CR Scientific and Compliance Consulting, LLC, Gainesville, FL, 32608, USA
| | - Andrew Specht
- Department of Small Animal Clinical Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| | - Kirsten E Coleman
- Powell Gene Therapy Center, University of Florida, Gainesville, FL, 32610, USA
| | - Laurie M Brown
- Glycogen Storage Disease Program, University of Florida College of Medicine, Gainesville, FL, USA
| | - Ana M Estrella
- Glycogen Storage Disease Program, Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
- Glycogen Storage Disease Program, University of Florida College of Medicine, Gainesville, FL, USA
| | - Monika Dambska
- Glycogen Storage Disease Program, University of Florida College of Medicine, Gainesville, FL, USA
- Glycogen Storage Disease Program, Connecticut Children's Medical Center, Hartford, CT, USA
| | - Kathryn R Dahlberg
- Glycogen Storage Disease Program, Connecticut Children's Medical Center, Hartford, CT, USA
| | - David A Weinstein
- Glycogen Storage Disease Program, Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT, 06030, USA.
- Glycogen Storage Disease Program, University of Florida College of Medicine, Gainesville, FL, USA.
- Glycogen Storage Disease Program, Connecticut Children's Medical Center, Hartford, CT, USA.
| |
Collapse
|
28
|
Bertinat R, Westermeier F, Gatica R, Nualart F. Sodium tungstate: Is it a safe option for a chronic disease setting, such as diabetes? J Cell Physiol 2018; 234:51-60. [DOI: 10.1002/jcp.26913] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 06/13/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Romina Bertinat
- Centro de Microscopía Avanzada, CMA Bio‐Bio Facultad de Ciencias Biológicas, Universidad de Concepción Concepción Chile
| | - Francisco Westermeier
- Department of Health Studies Institute of Biomedical Science, FH JOANNEUM Gesellschaft mbH University of Applied Sciences Graz Austria
- Facultad de Ciencia, Universidad San Sebastián Santiago Chile
| | - Rodrigo Gatica
- Laboratorio de Patología Veterinaria Escuela de Veterinaria, Facultad de Ciencias, Universidad Mayor Santiago Chile
| | - Francisco Nualart
- Centro de Microscopía Avanzada, CMA Bio‐Bio Facultad de Ciencias Biológicas, Universidad de Concepción Concepción Chile
| |
Collapse
|
29
|
Monteillet L, Gjorgjieva M, Silva M, Verzieux V, Imikirene L, Duchampt A, Guillou H, Mithieux G, Rajas F. Intracellular lipids are an independent cause of liver injury and chronic kidney disease in non alcoholic fatty liver disease-like context. Mol Metab 2018; 16:100-115. [PMID: 30100243 PMCID: PMC6157648 DOI: 10.1016/j.molmet.2018.07.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/19/2018] [Accepted: 07/23/2018] [Indexed: 12/15/2022] Open
Abstract
Objective Ectopic lipid accumulation in the liver and kidneys is a hallmark of metabolic diseases leading to non-alcoholic fatty liver disease (NAFLD) and chronic kidney disease (CKD). Moreover, recent data have highlighted a strong correlation between NAFLD and CKD incidences. In this study, we use two mouse models of hepatic steatosis or CKD, each initiated independently of the other upon the suppression of glucose production specifically in the liver or kidneys, to elucidate the mechanisms underlying the development of CKD in the context of NAFLD-like pathology. Methods Mice with a deletion of G6pc, encoding glucose-6 phosphatase catalytic subunit, specifically in the liver (L.G6pc−/− mice) or the kidneys (K.G6pc−/− mice), were fed with either a standard diet or a high fat/high sucrose (HF/HS) diet during 9 months. These mice represent two original models of a rare metabolic disease named Glycogen Storage Disease Type Ia (GSDIa) that is characterized by both NAFLD-like pathology and CKD. Two other groups of L.G6pc−/− and K.G6pc−/− mice were fed a standard diet for 6 months and then treated with fenofibrate for 3 months. Lipid and glucose metabolisms were characterized, and NAFLD-like and CKD damages were evaluated. Results Lipid depot exacerbation upon high-calorie diet strongly accelerated hepatic and renal pathologies induced by the G6pc-deficiency. In L.G6pc−/− mice, HF/HS diet increased liver injuries, characterized by higher levels of plasmatic transaminases and increased hepatic tumor incidence. In K.G6pc−/− mice, HF/HS diet increased urinary albumin and lipocalin 2 excretion and aggravated renal fibrosis. In both cases, the worsening of NAFLD-like injuries and CKD was independent of glycogen content. Furthermore, fenofibrate, via the activation of lipid oxidation significantly decreased the hepatic or renal lipid accumulations and prevented liver or kidney damages in L.G6pc−/− and K.G6pc−/− mice, respectively. Finally, we show that L.G6pc−/− mice and K.G6pc−/− mice developed NAFLD-like pathology and CKD independently. Conclusions This study highlights the crucial role that lipids play in the independent development of both NAFLD and CKD and demonstrates the importance of lipid-lowering treatments in various metabolic diseases featured by lipid load, from the “rare” GSDIa to the “epidemic” morbid obesity or type 2 diabetes. Exacerbating lipid accumulation aggravates liver/kidney injury in GSDI. Fenofibrate-mediated PPARα activation induces hepatic and renal lipid turnover. Increased lipid turnover prevents glycogen synthesis and accumulation. PPARα–mediated metabolic reprograming prevents hepatic and renal GSDI complications. NAFLD and CKD develop independently.
Collapse
Affiliation(s)
- Laure Monteillet
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Monika Gjorgjieva
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Marine Silva
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Vincent Verzieux
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Linda Imikirene
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Adeline Duchampt
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Hervé Guillou
- Toxalim, Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, 31027, France.
| | - Gilles Mithieux
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Fabienne Rajas
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| |
Collapse
|
30
|
Kaneko K, Soty M, Zitoun C, Duchampt A, Silva M, Philippe E, Gautier-Stein A, Rajas F, Mithieux G. The role of kidney in the inter-organ coordination of endogenous glucose production during fasting. Mol Metab 2018; 16:203-212. [PMID: 29960865 PMCID: PMC6157617 DOI: 10.1016/j.molmet.2018.06.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 06/04/2018] [Accepted: 06/13/2018] [Indexed: 11/15/2022] Open
Abstract
Objective The respective contributions to endogenous glucose production (EGP) of the liver, kidney and intestine vary during fasting. We previously reported that the deficiency in either hepatic or intestinal gluconeogenesis modulates the repartition of EGP via glucagon secretion (humoral factor) and gut–brain–liver axis (neural factor), respectively. Considering renal gluconeogenesis reportedly accounted for approximately 50% of EGP during fasting, we examined whether a reduction in renal gluconeogenesis could promote alterations in the repartition of EGP in this situation. Methods We studied mice whose glucose-6-phosphatase (G6Pase) catalytic subunit (G6PC) is specifically knocked down in the kidneys (K-G6pc-/- mice) during fasting. We also examined the additional effects of intestinal G6pc deletion, renal denervation and vitamin D administration on the altered glucose metabolism in K-G6pc-/- mice. Results Compared with WT mice, K-G6pc-/- mice exhibited (1) lower glycemia, (2) enhanced intestinal but not hepatic G6Pase activity, (3) enhanced hepatic glucokinase (GK encoded by Gck) activity, (4) increased hepatic glucose-6-phosphate and (5) hepatic glycogen spared from exhaustion during fasting. Increased hepatic Gck expression in the post-absorptive state could be dependent on the enhancement of insulin signal (AKT phosphorylation) in K-G6pc-/- mice. In contrast, the increase in hepatic GK activity was not observed in mice with both kidney- and intestine-knockout (KI-G6pc-/- mice). Hepatic Gck gene expression and hepatic AKT phosphorylation were reduced in KI-G6pc-/- mice. Renal denervation by capsaicin did not induce any effect on glucose metabolism in K-G6pc-/- mice. Plasma level of 1,25 (OH)2 D3, an active form of vitamin D, was decreased in K-G6pc-/- mice. Interestingly, the administration of 1,25 (OH)2 D3 prevented the enhancement of intestinal gluconeogenesis and hepatic GK activity and blocked the accumulation of hepatic glycogen otherwise observed in K-G6pc-/- mice during fasting. Conclusions A diminution in renal gluconeogenesis that is accompanied by a decrease in blood vitamin D promotes a novel repartition of EGP among glucose producing organs during fasting, featured by increased intestinal gluconeogenesis that leads to sparing glycogen stores in the liver. Our data suggest a possible involvement of a crosstalk between the kidneys and intestine (via the vitamin D system) and the intestine and liver (via a neural gut-brain axis), which might take place in the situations of deficient renal glucose production, such as chronic kidney disease. Reduced renal G6Pase activity promotes increased hepatic glycogen during fasting. Reduced renal G6Pase activity enhances intestinal but not hepatic G6Pase activity. Reduced renal G6Pase activity results in low vitamin D level. Vitamin D injection restores metabolism in mice with reduced renal G6Pase activity.
Collapse
Affiliation(s)
- Keizo Kaneko
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Maud Soty
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France
| | - Carine Zitoun
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France
| | - Adeline Duchampt
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France
| | - Marine Silva
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France
| | - Erwann Philippe
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France
| | - Amandine Gautier-Stein
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France
| | - Fabienne Rajas
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France
| | - Gilles Mithieux
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| |
Collapse
|
31
|
Bertinat R, Westermeier F, Silva P, Gatica R, Oliveira JM, Nualart F, Gomis R, Yáñez AJ. The Antidiabetic Agent Sodium Tungstate Induces Abnormal Glycogen Accumulation in Renal Proximal Tubules from Diabetic IRS2-Knockout Mice. J Diabetes Res 2018; 2018:5697970. [PMID: 30003110 PMCID: PMC5996472 DOI: 10.1155/2018/5697970] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/19/2017] [Accepted: 01/28/2018] [Indexed: 11/22/2022] Open
Abstract
The kidney is an insulin-sensitive organ involved in glucose homeostasis. One major effect of insulin is to induce glycogen storage in the liver and muscle. However, no significant glycogen stores are detected in normal kidneys, but diabetic subjects present a characteristic renal histopathological feature resulting from extensive glycogen deposition mostly in nonproximal tubules. The mechanism of renal glycogen accumulation is yet poorly understood. Here, we studied in situ glycogen accumulation in the kidney from diabetic IRS2-knockout mice and the effect of the insulin-mimetic agent sodium tungstate (NaW). IRS2-knockout mice displayed hyperglycemia and hyperinsulinemia. NaW only normalized glycemia. There was no evident morphological difference between kidneys from untreated wild-type (WT), NaW-treated WT, and untreated IRS2-knockout mice. However, NaW-treated IRS2-knockout mice showed tubular alterations resembling clear cells in the cortex, but not in the outer medulla, that were correlated with glycogen accumulation. Immunohistochemical detection of the gluconeogenic enzyme phosphoenolpyruvate carboxykinase, mostly expressed by renal proximal tubules, showed that altered tubules were of proximal origin. Our preliminary study suggests that IRS2 differentially regulates glycogen accumulation in renal tubules and that NaW treatment in the context of IRS2 ablation induces abnormal glycogen accumulation in cortical proximal tubules.
Collapse
Affiliation(s)
- Romina Bertinat
- Centro de Microscopía Avanzada (CMA BIO-BIO), Universidad de Concepción, Concepción, Chile
| | - Francisco Westermeier
- Institute of Biomedical Science, FH Joanneum Gesellschaft mbH University of Applied Sciences, Eggenberger Allee 13, 8020 Graz, Austria
- Facultad de Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Pamela Silva
- Facultad de Salud, Universidad Santo Tomás, Osorno, Chile
| | - Rodrigo Gatica
- Escuela de Veterinaria, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Joana Moitinho Oliveira
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
- Diabetes and Obesity Research Laboratory, IDIBAPS, Barcelona, Spain
| | - Francisco Nualart
- Centro de Microscopía Avanzada (CMA BIO-BIO), Universidad de Concepción, Concepción, Chile
| | - Ramón Gomis
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
- Diabetes and Obesity Research Laboratory, IDIBAPS, Barcelona, Spain
- Department of Endocrinology and Nutrition, Hospital Clinic, Barcelona, Spain
- Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | | |
Collapse
|
32
|
Bontha SV, Maluf DG, Archer KJ, Dumur CI, Dozmorov M, King A, Akalin E, Mueller TF, Gallon L, Mas VR. Effects of DNA Methylation on Progression to Interstitial Fibrosis and Tubular Atrophy in Renal Allograft Biopsies: A Multi-Omics Approach. Am J Transplant 2017; 17:3060-3075. [PMID: 28556588 PMCID: PMC5734859 DOI: 10.1111/ajt.14372] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 05/01/2017] [Accepted: 05/20/2017] [Indexed: 01/25/2023]
Abstract
Progressive fibrosis of the interstitium is the dominant final pathway in renal destruction in native and transplanted kidneys. Over time, the continuum of molecular events following immunological and nonimmunological insults lead to interstitial fibrosis and tubular atrophy and culminate in kidney failure. We hypothesize that these insults trigger changes in DNA methylation (DNAm) patterns, which in turn could exacerbate injury and slow down the regeneration processes, leading to fibrosis development and graft dysfunction. Herein, we analyzed biopsy samples from kidney allografts collected 24 months posttransplantation and used an integrative multi-omics approach to understand the underlying molecular mechanisms. The role of DNAm and microRNAs on the graft gene expression was evaluated. Enrichment analyses of differentially methylated CpG sites were performed using GenomeRunner. CpGs were strongly enriched in regions that were variably methylated among tissues, implying high tissue specificity in their regulatory impact. Corresponding to this methylation pattern, gene expression data were related to immune response (activated state) and nephrogenesis (inhibited state). Preimplantation biopsies showed similar DNAm patterns to normal allograft biopsies at 2 years posttransplantation. Our findings demonstrate for the first time a relationship among epigenetic modifications and development of interstitial fibrosis, graft function, and inter-individual variation on long-term outcomes.
Collapse
Affiliation(s)
- Sai Vineela Bontha
- Translational Genomics Transplant Laboratory, Transplant Division, University of Virginia, Department of Surgery, PO Box 800625. 409 Lane Rd, Charlottesville, VA, 22908- 0625, USA
| | - Daniel G. Maluf
- Translational Genomics Transplant Laboratory, Transplant Division, University of Virginia, Department of Surgery, PO Box 800625. 409 Lane Rd, Charlottesville, VA, 22908- 0625, USA
| | - Kellie J. Archer
- Division of Biostatistics, The Ohio State University, 1841 Neil Avenue, 240 Cunz Hall, Columbus, OH 43210
| | - Catherine I. Dumur
- Department of Pathology, Virginia Commonwealth University, PO Box 980662, 1101 E. Marshall Street, Richmond, VA 23298-0662
| | - Mikhail Dozmorov
- Department of Biostatistics, Virginia Commonwealth University, One Capitol Square, room 730, 830 East Main Street, Richmond, Virginia 23298
| | - Anne King
- Division of Nephrology, Internal Medicine. Virginia commonwealth University, VA, 1101 E. Marshall Street, Richmond, VA 23298-0662
| | - Enver Akalin
- Departments of Clinical Medicine and Surgery, Albert Einstein College of Medicine Montefiore Medical Center, 11 E 210th St, Bronx, NY 10467
| | - Thomas F. Mueller
- Division of Nephorology, Internal Medicine, University Hospital Zurich, Ramistrasse 100, Zurich-8091
| | - Lorenzo Gallon
- Department of Medicine-Nephrology, Northwestern University676 N St Clair St # 100, Chicago, IL 60611
| | - Valeria R. Mas
- Translational Genomics Transplant Laboratory, Transplant Division, University of Virginia, Department of Surgery, PO Box 800625. 409 Lane Rd, Charlottesville, VA, 22908- 0625, USA
| |
Collapse
|
33
|
Hijmans BS, Boss A, van Dijk TH, Soty M, Wolters H, Mutel E, Groen AK, Derks TGJ, Mithieux G, Heerschap A, Reijngoud DJ, Rajas F, Oosterveer MH. Hepatocytes contribute to residual glucose production in a mouse model for glycogen storage disease type Ia. Hepatology 2017; 66:2042-2054. [PMID: 28727166 DOI: 10.1002/hep.29389] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 07/02/2017] [Accepted: 07/15/2017] [Indexed: 01/14/2023]
Abstract
UNLABELLED It is a long-standing enigma how glycogen storage disease (GSD) type I patients retain a limited capacity for endogenous glucose production despite the loss of glucose-6-phosphatase activity. Insight into the source of residual endogenous glucose production is of clinical importance given the risk of sudden death in these patients, but so far contradictory mechanisms have been proposed. We investigated glucose-6-phosphatase-independent endogenous glucose production in hepatocytes isolated from a liver-specific GSD Ia mouse model (L-G6pc-/- mice) and performed real-time analysis of hepatic glucose fluxes and glycogen metabolism in L-G6pc-/- mice using state-of-the-art stable isotope methodologies. Here we show that G6pc-deficient hepatocytes are capable of producing glucose. In vivo analysis of hepatic glucose metabolism revealed that the hepatic glucokinase flux was decreased by 95% in L-G6pc-/- mice. It also showed increased glycogen phosphorylase flux in L-G6pc-/- mice, which is coupled to the release of free glucose through glycogen debranching. Although the ex vivo activities of debranching enzyme and lysosomal acid maltase, two major hepatic α-glucosidases, were unaltered in L-G6pc-/- mice, pharmacological inhibition of α-glucosidase activity almost completely abolished residual glucose production by G6pc-deficient hepatocytes. CONCLUSION Our data indicate that hepatocytes contribute to residual glucose production in GSD Ia. We show that α-glucosidase activity, i.e. glycogen debranching and/or lysosomal glycogen breakdown, contributes to residual glucose production by GSD Ia hepatocytes. A strong reduction in hepatic GCK flux in L-G6pc-/- mice furthermore limits the phosphorylation of free glucose synthesized by G6pc-deficient hepatocytes, allowing the release of glucose into the circulation. The almost complete abrogation of GCK flux in G6pc-deficient liver also explains the contradictory reports on residual glucose production in GSD Ia patients. (Hepatology 2017;66:2042-2054).
Collapse
Affiliation(s)
- Brenda S Hijmans
- Department of Pediatrics, Center for Liver Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Andreas Boss
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Theo H van Dijk
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Maud Soty
- Institut National de la Santé et de la Recherche Médicale U1213.,Université de Lyon, Lyon, France.,Université Lyon 1, Lyon, France
| | - Henk Wolters
- Department of Pediatrics, Center for Liver Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Elodie Mutel
- Institut National de la Santé et de la Recherche Médicale U1213.,Université de Lyon, Lyon, France.,Université Lyon 1, Lyon, France
| | - Albert K Groen
- Department of Pediatrics, Center for Liver Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Terry G J Derks
- Section of Metabolic Diseases, Beatrix Children's Hospital, Center for Liver Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gilles Mithieux
- Institut National de la Santé et de la Recherche Médicale U1213.,Université de Lyon, Lyon, France.,Université Lyon 1, Lyon, France
| | - Arend Heerschap
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dirk-Jan Reijngoud
- Department of Pediatrics, Center for Liver Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Fabienne Rajas
- Institut National de la Santé et de la Recherche Médicale U1213.,Université de Lyon, Lyon, France.,Université Lyon 1, Lyon, France
| | - Maaike H Oosterveer
- Department of Pediatrics, Center for Liver Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
34
|
Chou JY, Kim GY, Cho JH. Recent development and gene therapy for glycogen storage disease type Ia. LIVER RESEARCH 2017; 1:174-180. [PMID: 29576889 PMCID: PMC5859325 DOI: 10.1016/j.livres.2017.12.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Glycogen storage disease type Ia (GSD-Ia) is an autosomal recessive metabolic disorder caused by a deficiency in glucose-6-phosphatase-α (G6Pase-α or G6PC) that is expressed primarily in the liver, kidney, and intestine. G6Pase-α catalyzes the hydrolysis of glucose-6-phosphate (G6P) to glucose and phosphate in the terminal step of gluconeogenesis and glycogenolysis, and is a key enzyme for endogenous glucose production. The active site of G6Pase-α is inside the endoplasmic reticulum (ER) lumen. For catalysis, the substrate G6P must be translocated from the cytoplasm into the ER lumen by a G6P transporter (G6PT). The functional coupling of G6Pase-α and G6PT maintains interprandial glucose homeostasis. Dietary therapies for GSD-Ia are available, but cannot prevent the long-term complication of hepatocellular adenoma that may undergo malignant transformation to hepatocellular carcinoma. Animal models of GSD-Ia are now available and are being exploited to both delineate the disease more precisely and develop new treatment approaches, including gene therapy.
Collapse
Affiliation(s)
- Janice Y. Chou
- Section on Cellular Differentiation, Eunice Kennedy Shriver National
Institute of Child Health and Human Development, National Institutes of Health,
Bethesda, MD, USA
| | - Goo-Young Kim
- Section on Cellular Differentiation, Eunice Kennedy Shriver National
Institute of Child Health and Human Development, National Institutes of Health,
Bethesda, MD, USA
| | - Jun-Ho Cho
- Section on Cellular Differentiation, Eunice Kennedy Shriver National
Institute of Child Health and Human Development, National Institutes of Health,
Bethesda, MD, USA
| |
Collapse
|
35
|
Abdul-Wahed A, Guilmeau S, Postic C. Sweet Sixteenth for ChREBP: Established Roles and Future Goals. Cell Metab 2017; 26:324-341. [PMID: 28768172 DOI: 10.1016/j.cmet.2017.07.004] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 06/01/2017] [Accepted: 07/12/2017] [Indexed: 12/25/2022]
Abstract
With the identification of ChREBP in 2001, our interest in understanding the molecular control of carbohydrate sensing has surged. While ChREBP was initially studied as a master regulator of lipogenesis in liver and fat tissue, it is now clear that ChREBP functions as a central metabolic coordinator in a variety of cell types in response to environmental and hormonal signals, with wide implications in health and disease. Celebrating its sweet sixteenth birthday, we review here the current knowledge about the function and regulation of ChREBP throughout usual and less explored tissues, to recapitulate ChREBP's role as a whole-body glucose sensor.
Collapse
Affiliation(s)
- Aya Abdul-Wahed
- Inserm, U1016, Institut Cochin, 75014 Paris, France; CNRS UMR 8104, 75014 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Sandra Guilmeau
- Inserm, U1016, Institut Cochin, 75014 Paris, France; CNRS UMR 8104, 75014 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Catherine Postic
- Inserm, U1016, Institut Cochin, 75014 Paris, France; CNRS UMR 8104, 75014 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France.
| |
Collapse
|
36
|
Gjorgjieva M, Oosterveer MH, Mithieux G, Rajas F. Mechanisms by Which Metabolic Reprogramming in GSD1 Liver Generates a Favorable Tumorigenic Environment. JOURNAL OF INBORN ERRORS OF METABOLISM AND SCREENING 2016. [DOI: 10.1177/2326409816679429] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Monika Gjorgjieva
- Institut National de la Santé et de la Recherche Médicale, U1213 “Nutrition, Diabetes and the Brain”, Lyon, France
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
| | - Maaike H. Oosterveer
- Department of Pediatrics, Center for Liver, Digestive, and Metabolic Diseases, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Gilles Mithieux
- Institut National de la Santé et de la Recherche Médicale, U1213 “Nutrition, Diabetes and the Brain”, Lyon, France
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
| | - Fabienne Rajas
- Institut National de la Santé et de la Recherche Médicale, U1213 “Nutrition, Diabetes and the Brain”, Lyon, France
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
| |
Collapse
|
37
|
Rines AK, Sharabi K, Tavares CDJ, Puigserver P. Targeting hepatic glucose metabolism in the treatment of type 2 diabetes. Nat Rev Drug Discov 2016; 15:786-804. [PMID: 27516169 DOI: 10.1038/nrd.2016.151] [Citation(s) in RCA: 232] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Type 2 diabetes mellitus is characterized by the dysregulation of glucose homeostasis, resulting in hyperglycaemia. Although current diabetes treatments have exhibited some success in lowering blood glucose levels, their effect is not always sustained and their use may be associated with undesirable side effects, such as hypoglycaemia. Novel antidiabetic drugs, which may be used in combination with existing therapies, are therefore needed. The potential of specifically targeting the liver to normalize blood glucose levels has not been fully exploited. Here, we review the molecular mechanisms controlling hepatic gluconeogenesis and glycogen storage, and assess the prospect of therapeutically targeting associated pathways to treat type 2 diabetes.
Collapse
Affiliation(s)
- Amy K Rines
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Kfir Sharabi
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Clint D J Tavares
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Pere Puigserver
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
38
|
Gjorgjieva M, Raffin M, Duchampt A, Perry A, Stefanutti A, Brevet M, Tortereau A, Dubourg L, Hubert-Buron A, Mabille M, Pelissou C, Lassalle L, Labrune P, Mithieux G, Rajas F. Progressive development of renal cysts in glycogen storage disease type I. Hum Mol Genet 2016; 25:3784-3797. [PMID: 27436577 DOI: 10.1093/hmg/ddw224] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 06/15/2016] [Accepted: 06/15/2016] [Indexed: 01/25/2023] Open
Abstract
Glycogen storage disease type I (GSDI) is a rare metabolic disease due to glucose-6 phosphatase deficiency, characterized by fasting hypoglycemia. Patients also develop chronic kidney disease whose mechanisms are poorly understood. To decipher the process, we generated mice with a kidney-specific knockout of glucose-6 phosphatase (K.G6pc-/- mice) that exhibited the first signs of GSDI nephropathy after 6 months of G6pc deletion. We studied the natural course of renal deterioration in K.G6pc-/- mice for 18 months and observed the progressive deterioration of renal functions characterized by early tubular dysfunction and a later destruction of the glomerular filtration barrier. After 15 months, K.G6pc-/- mice developed tubular-glomerular fibrosis and podocyte injury, leading to the development of cysts and renal failure. On the basis of these findings, we were able to detect the development of cysts in 7 out of 32 GSDI patients, who developed advanced renal impairment. Of these 7 patients, 3 developed renal failure. In addition, no renal cysts were detected in six patients who showed early renal impairment. In conclusion, renal pathology in GSDI is characterized by progressive tubular dysfunction and the development of polycystic kidneys that probably leads to the development of irreversible renal failure in the late stages. Systematic observations of cyst development by kidney imaging should improve the evaluation of the disease's progression, independently of biochemical markers.
Collapse
Affiliation(s)
- Monika Gjorgjieva
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, France.,Université de Lyon, Lyon, France.,Université Lyon1, Villeurbanne, France
| | - Margaux Raffin
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, France.,Université de Lyon, Lyon, France.,Université Lyon1, Villeurbanne, France
| | - Adeline Duchampt
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, France.,Université de Lyon, Lyon, France.,Université Lyon1, Villeurbanne, France
| | - Ariane Perry
- APHP, Hôpitaux Universitaires Paris Sud, Hôpital Antoine Béclère, Centre de référence des maladies héréditaires du métabolisme hépatique, Clamart, France
| | - Anne Stefanutti
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, France.,Université de Lyon, Lyon, France.,Université Lyon1, Villeurbanne, France
| | - Marie Brevet
- Université de Lyon, Lyon, France.,Service de pathologie et de neuropathologie Est, Institut de Cancérologie des Hospices Civils de Lyon, Lyon, France
| | - Antonin Tortereau
- Université de Lyon, Lyon, France.,VetAgro Sup, UPSP 2011-03-101, ICE, Marcy L'Etoile, France
| | - Laurence Dubourg
- Université de Lyon, Lyon, France.,Université Lyon1, Villeurbanne, France.,UMR 5305 CNRS/Université Claude-Bernard, Biologie tissulaire et Ingénierie thérapeutique, Lyon, France.,Exploration Fonctionnelle Rénale, Groupement Hospitalier Edouard Herriot, Hospices civils de Lyon, Lyon, France
| | - Aurélie Hubert-Buron
- APHP, Hôpitaux Universitaires Paris Sud, Hôpital Antoine Béclère, Centre de référence des maladies héréditaires du métabolisme hépatique, Clamart, France
| | - Mylène Mabille
- APHP, Hôpitaux Universitaires Paris Sud, Hôpital Antoine Béclère, Service de radiologie, Clamart, France.,Université Paris Sud, Orsay, France
| | - Coralie Pelissou
- APHP, Hôpitaux Universitaires Paris Sud, Hôpital Antoine Béclère, Service de radiologie, Clamart, France.,Université Paris Sud, Orsay, France
| | - Louis Lassalle
- APHP, Hôpitaux Universitaires Paris Sud, Hôpital Antoine Béclère, Service de radiologie, Clamart, France.,Université Paris Sud, Orsay, France
| | - Philippe Labrune
- APHP, Hôpitaux Universitaires Paris Sud, Hôpital Antoine Béclère, Centre de référence des maladies héréditaires du métabolisme hépatique, Clamart, France.,Université Paris Sud, Orsay, France
| | - Gilles Mithieux
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, France.,Université de Lyon, Lyon, France.,Université Lyon1, Villeurbanne, France
| | - Fabienne Rajas
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, France .,Université de Lyon, Lyon, France.,Université Lyon1, Villeurbanne, France
| |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW Glycogen storage disorders (GSDs) are inborn errors of metabolism with abnormal storage or utilization of glycogen. The present review focuses on recent advances in hepatic GSD types I, III and VI/IX, with emphasis on clinical aspects and treatment. RECENT FINDINGS Evidence accumulates that poor metabolic control is a risk factor for the development of long-term complications, such as liver adenomas, low bone density/osteoporosis, and kidney disease in GSD I. However, mechanisms leading to these complications remain poorly understood and are being investigated. Molecular causes underlying neutropenia and neutrophil dysfunction in GSD I have been elucidated. Case series provide new insights into the natural course and outcome of GSD types VI and IX. For GSD III, a high protein/fat diet has been reported to improve (cardio)myopathy, but the beneficial effect of this dietary concept on muscle and liver disease manifestations needs to be further established in prospective studies. SUMMARY Although further knowledge has been gained regarding pathophysiology, disease course, treatment, and complications of hepatic GSDs, more controlled prospective studies are needed to assess effects of different dietary and medical treatment options on long-term outcome and quality of life.
Collapse
Affiliation(s)
- Patricie Burda
- aDivision of Metabolism and Children's Research Center, University Children's Hospital bDivision of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich cradiz - Rare Disease Initiative Zurich, Clinical Research Priority Program for Rare Diseases, University of Zurich, Switzerland
| | | |
Collapse
|
40
|
Ben Chehida A, Bensmaïl T, Ben Rehouma F, Ben Abdelaziz R, Azzouz H, Boudabbous H, Slim Abdelmoula M, Abdelhak S, Kaabachi N, Ben Turkia H, Tebib N. [Renal involvement in glycogen storage disease type 1: Practical issues]. Nephrol Ther 2015; 11:240-5. [PMID: 25957470 DOI: 10.1016/j.nephro.2014.12.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Revised: 12/26/2014] [Accepted: 12/30/2014] [Indexed: 11/27/2022]
Abstract
AIM To investigate risk factors of renal complications in glycogen storage disease type I, in order to identify practical implications for renal preservation. METHODS A retrospective study of 38 patients with glycogen storage disease type I. RESULTS The patients studied were 8.6 years old in average (1.5 to 22 years) and were followed during 7.4 ± 4.5 years. Hypercalciuria was detected in 23 patients and was related to acidosis (P=0.028), higher lactate levels (5.9 ± 3.5 versus 3.7 ± 1.7 mmol/L; P=0.013) and smaller height (-2.1 ± 1.5 SD versus -0.8 ± 1.5 SD; P=0.026). Urolithiasis was diagnosed in 7 cases. Glomerular disease (19/38) was more frequent in cases with severe hypertriglyceridemia (P=0.042) and occurred at an older age (P=0.007). Microalbuminuria occurred in 15/31 cases; ACE inhibitors were prescribed in only 8 cases. The frequency of renal complications did not differ according to the diet group (continuous enteral feeding or uncooked starch). Logistic regression concluded as risk factors: lactic acidosis for tubular disease and age>10 years for glomerular disease. CONCLUSIONS Renal involvement is common in glycogen storage disease type I patients. Tubular abnormalities are precocious, related to lactic acidosis and may be detected by monitoring of urinary calcium. Glomerular hyperfiltration is the first stage of a progressive glomerular disease and is related to age. Practical implications for renal preservation are discussed based on our results and literature.
Collapse
Affiliation(s)
- Amel Ben Chehida
- Service de pédiatrie et maladies métaboliques héréditaires, hôpital la Rabta, Jabberi, 1007 Tunis, Tunisie.
| | - Takoua Bensmaïl
- Service de pédiatrie et maladies métaboliques héréditaires, hôpital la Rabta, Jabberi, 1007 Tunis, Tunisie
| | - Faten Ben Rehouma
- Laboratoire de génomique biomédicale et oncogénétique, institut Pasteur de Tunis, 13, place Pasteur, 1002 Tunis, Tunisie
| | - Rim Ben Abdelaziz
- Service de pédiatrie et maladies métaboliques héréditaires, hôpital la Rabta, Jabberi, 1007 Tunis, Tunisie
| | - Hatem Azzouz
- Service de pédiatrie et maladies métaboliques héréditaires, hôpital la Rabta, Jabberi, 1007 Tunis, Tunisie
| | - Hela Boudabbous
- Service de pédiatrie et maladies métaboliques héréditaires, hôpital la Rabta, Jabberi, 1007 Tunis, Tunisie
| | - Mohamed Slim Abdelmoula
- Service de pédiatrie et maladies métaboliques héréditaires, hôpital la Rabta, Jabberi, 1007 Tunis, Tunisie
| | - Sonia Abdelhak
- Laboratoire de génomique biomédicale et oncogénétique, institut Pasteur de Tunis, 13, place Pasteur, 1002 Tunis, Tunisie
| | - Naziha Kaabachi
- Laboratoire de biochimie, hôpital la Rabta, Jabberi, 1007 Tunis, Tunisie
| | - Hadhami Ben Turkia
- Service de pédiatrie et maladies métaboliques héréditaires, hôpital la Rabta, Jabberi, 1007 Tunis, Tunisie
| | - Néji Tebib
- Service de pédiatrie et maladies métaboliques héréditaires, hôpital la Rabta, Jabberi, 1007 Tunis, Tunisie
| |
Collapse
|
41
|
Rajas F, Clar J, Gautier-Stein A, Mithieux G. Lessons from new mouse models of glycogen storage disease type 1a in relation to the time course and organ specificity of the disease. J Inherit Metab Dis 2015; 38:521-7. [PMID: 25164786 PMCID: PMC5522669 DOI: 10.1007/s10545-014-9761-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Revised: 08/05/2014] [Accepted: 08/07/2014] [Indexed: 12/12/2022]
Abstract
Patients with glycogen storage diseases type 1 (GSD1) suffer from life-threatening hypoglycaemia, when left untreated. Despite an intensive dietary treatment, patients develop severe complications, such as liver tumors and renal failure, with aging. Until now, the animal models available for studying the GSD1 did not survive after weaning. To gain further insights into the molecular mechanisms of the disease and to evaluate potential treatment strategies, we have recently developed novel mouse models in which the catalytic subunit of glucose-6 phosphatase (G6pc) is deleted in each glucose-producing organ specifically. For that, B6.G6pc(ex3lox/ex3lox) mice were crossed with transgenic mice expressing a recombinase under the control of the serum albumin, the kidney androgen protein or the villin promoter, in order to obtain liver, kidney or intestine G6pc(-/-) mice, respectively. As opposed to total G6pc knockout mice, tissue-specific G6pc deficiency allows mice to maintain their blood glucose by inducing glucose production in the other gluconeogenic organs. Even though it is considered that glucose is produced mainly by the liver, liver G6pc(-/-) mice are perfectly viable and exhibit the same hepatic pathological features as GSD1 patients, including the late development of hepatocellular adenomas and carcinomas. Interestingly, renal G6pc(-/-) mice developed renal symptoms similar to the early human GSD1 nephropathy. This includes glycogen overload that leads to nephromegaly and morphological and functional alterations in the kidneys. Thus, our data suggest that renal G6Pase deficiency per se is sufficient to induce the renal pathology of GSD1. Therefore, these new mouse models should allow us to improve the strategies of treatment on both nutritional and pharmacological points of view.
Collapse
Affiliation(s)
- Fabienne Rajas
- Institut National de la Santé et de la Recherche Médicale, U855, Lyon, 69008, France,
| | | | | | | |
Collapse
|