1
|
Siri B, Greco B, Martinelli D, Cairoli S, Guarnera A, Longo D, Napolitano A, Parrillo C, Ravà L, Simeoli R, Spagnoletti G, Taurisano R, Veraldi S, Pietrobattista A, Spada M, Dionisi‐Vici C. Positive Clinical, Neuropsychological, and Metabolic Impact of Liver Transplantation in Patients With Argininosuccinate Lyase Deficiency. J Inherit Metab Dis 2025; 48:e12843. [PMID: 39776112 PMCID: PMC11706762 DOI: 10.1002/jimd.12843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/03/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025]
Abstract
Liver transplantation (LTx) is increasingly used in Urea Cycle Defects (UCDs) to prevent recurrent hyperammonemia and related neurological irreversible injury. Among UCDs, argininosuccinate lyase deficiency (ASLD) has a more complex phenotype than other UCDs, with long-term neurocognitive deficits. Therefore, the role of LTx in ASLD is still debated. The impact of LTx on nine patients with early-onset ASLD was assessed through pre- and post-LTx clinical, neuropsychological, MRI and biochemical evaluations. After LTx, no episodes of metabolic decompensations were reported. Neuropsychological evaluations documented significant improvement in cognitive/developmental functioning especially in patients transplanted in early childhood. Improvements were also highlighted in daily living skills and emotional-behavioral problems, with a reduction in attention disturbances and somatic complaints. Movement disorders resolved after LTx in patient transplanted in early childhood. Any patients developed epilepsy with stability of EEG alterations after LTx. A positive effect of LTx on other disease-related outcomes such as growth, diet, medications, hospitalizations, and long-term ASLD-related complications was highlighted. The primary biomarker argininosuccinic acid dramatically reduced in plasma after transplantation with a decreasing trend in CSF at long-term follow-up. Moreover, health-related quality of life improved after LTx, especially when assessed through MetabQoL, a tool designed for intoxication diseases such as ASLD. In conclusion, our study showed a global beneficial impact of LTx in early-onset ASLD patients to avoid episodes of hyperammonemia, and improve neurocognitive outcome, adaptive and behavioral deficits when performed in early childhood with a dramatic benefit in terms of quality of life.
Collapse
Affiliation(s)
- Barbara Siri
- Division of Metabolic Diseases and HepatologyBambino Gesù Children's Hospital IRCCSRomeItaly
| | - Benedetta Greco
- Division of Metabolic Diseases and HepatologyBambino Gesù Children's Hospital IRCCSRomeItaly
| | - Diego Martinelli
- Division of Metabolic Diseases and HepatologyBambino Gesù Children's Hospital IRCCSRomeItaly
| | - Sara Cairoli
- Division of Metabolic Diseases and HepatologyBambino Gesù Children's Hospital IRCCSRomeItaly
| | - Alessia Guarnera
- Neuroradiology Unit, Imaging DepartmentBambino Gesù Children's Hospital, IRCCSRomeItaly
| | - Daniela Longo
- Neuroradiology Unit, Imaging DepartmentBambino Gesù Children's Hospital, IRCCSRomeItaly
| | - Antonio Napolitano
- Medical Physics Unit, Risk Management Enterprise, Bambino Gesù Children's Hospital, IRCCSRomeItaly
| | - Chiara Parrillo
- Medical Physics Unit, Risk Management Enterprise, Bambino Gesù Children's Hospital, IRCCSRomeItaly
| | - Lucilla Ravà
- Epidemiology, Clinical Pathways and Clinical Risk Unit, Bambino Gesù Children's Hospital, IRCCSRomeItaly
| | - Raffaele Simeoli
- Division of Metabolic Diseases and HepatologyBambino Gesù Children's Hospital IRCCSRomeItaly
| | - Gionata Spagnoletti
- Division of Hepatobiliopancreatic Surgery, Liver and Kidney TransplantationBambino Gesù Children's Hospital, IRCCSRomeItaly
| | - Roberta Taurisano
- Division of Metabolic Diseases and HepatologyBambino Gesù Children's Hospital IRCCSRomeItaly
| | - Silvio Veraldi
- Division of Metabolic Diseases and HepatologyBambino Gesù Children's Hospital IRCCSRomeItaly
| | - Andrea Pietrobattista
- Division of Metabolic Diseases and HepatologyBambino Gesù Children's Hospital IRCCSRomeItaly
| | - Marco Spada
- Division of Hepatobiliopancreatic Surgery, Liver and Kidney TransplantationBambino Gesù Children's Hospital, IRCCSRomeItaly
| | - Carlo Dionisi‐Vici
- Division of Metabolic Diseases and HepatologyBambino Gesù Children's Hospital IRCCSRomeItaly
| |
Collapse
|
2
|
Patterson C, Gold A, So S, Kahnami L, Dworsky‐Fried M, Mamak E, Rogers A, Schulze A, Ertl‐Wagner B, Ng V, Avitzur Y. Long-term neurodevelopmental outcomes following liver transplantation for metabolic disease-a single centre experience. J Inherit Metab Dis 2025; 48:e12785. [PMID: 39135350 PMCID: PMC11670442 DOI: 10.1002/jimd.12785] [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: 02/28/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 12/28/2024]
Abstract
This study describes the neurodevelopmental outcome of children with urea cycle disorders (UCD) and organic acidemias (OA) preliver transplant (LT), 1-year, and 3-years post-LT. We performed a retrospective chart review of children with OA or UCD transplanted between January 2014 and December 2021. Standardized motor and cognitive assessment scores were collected from children who had ≥1 motor/cognitive assessment at any timepoint. Pre-LT brain magnetic resonance imaging (MRI) was graded. Associations between demographic/medical variables and neurodevelopmental outcomes were explored. Twenty-six children (64% male) underwent LT at a median age of 1.4 (interquartile range 0.71, 3.84) years. Fifteen (58%) had a UCD diagnosis, 14 (54%) required dialysis for hyperammonemia, and 10 (42%) had seizures typically around diagnosis. The proportion of children with gross motor scores >1 standard deviation (SD) below the mean increased across timepoints, and ≥50% demonstrated general intellect scores >2 SD below the mean at each timepoint. The following significant associations were noted: UCD diagnoses with lower general intellect scores (p = 0.019); arginosuccinate lyase deficiency diagnosis with lower visual motor scores at 3-years post-LT (p = 0.035); a history of seizures pre-LT with lower general intellect (>2SD below the mean) at 3-years post-LT (p = 0.020); dialysis pre-LT with lower motor scores (>1 SD below the mean) at 1-year post-LT (p = 0.039); pre-emptive LT with higher general intellect scores at 3-years post-LT (p = 0.001). MRI gradings were not associated with developmental scores. In our single centre study, children with UCD or OA had a higher prevalence of developmental impairment post-LT compared to population norms. Earlier screening, pre-emptive transplant, and rehabilitation may optimize long-term outcomes.
Collapse
Affiliation(s)
- Catherine Patterson
- Department of Rehabilitation ServicesThe Hospital for Sick ChildrenTorontoOntarioCanada
- Department of Physical TherapyUniversity of TorontoTorontoOntarioCanada
| | - Anna Gold
- Department of PsychologyThe Hospital for Sick ChildrenTorontoOntarioCanada
| | - Stephanie So
- Department of Rehabilitation ServicesThe Hospital for Sick ChildrenTorontoOntarioCanada
- Department of Physical TherapyUniversity of TorontoTorontoOntarioCanada
| | - Leila Kahnami
- Department of PsychologyThe Hospital for Sick ChildrenTorontoOntarioCanada
- Department of PsychologyYork UniversityTorontoOntarioCanada
| | - Michaela Dworsky‐Fried
- Department of PsychologyThe Hospital for Sick ChildrenTorontoOntarioCanada
- Department of PsychologyUniversity of OttawaOttawaOntarioCanada
| | - Eva Mamak
- Department of PsychologyThe Hospital for Sick ChildrenTorontoOntarioCanada
| | - Alaine Rogers
- Department of Rehabilitation ServicesThe Hospital for Sick ChildrenTorontoOntarioCanada
- Department of Physical TherapyUniversity of TorontoTorontoOntarioCanada
- Division of Clinical and Metabolic GeneticsThe Hospital for Sick ChildrenTorontoOntarioCanada
| | - Andreas Schulze
- Department of Occupational TherapyUniversity of TorontoTorontoOntarioCanada
- Department of Pediatrics and BiochemistryUniversity of TorontoTorontoOntarioCanada
| | - Birgit Ertl‐Wagner
- Department of Diagnostic and Interventional RadiologyHospital for Sick ChildrenTorontoOntarioCanada
- Department of Medical ImagingUniversity of TorontoTorontoOntarioCanada
| | - Vicky Ng
- Division of Gastroenterology, Hepatology and NutritionThe Hospital for Sick ChildrenTorontoOntarioCanada
- Department of Pediatrics, University of TorontoTorontoOntarioCanada
| | - Yaron Avitzur
- Division of Gastroenterology, Hepatology and NutritionThe Hospital for Sick ChildrenTorontoOntarioCanada
- Department of Pediatrics, University of TorontoTorontoOntarioCanada
| |
Collapse
|
3
|
Perry AS, Hadad N, Chatterjee E, Jimenez-Ramos M, Farber-Eger E, Roshani R, Stolze LK, Betti MJ, Zhao S, Huang S, Martens L, Kendall TJ, Thone T, Amancherla K, Bailin S, Gabriel CL, Koethe J, Carr JJ, Terry JG, Vaitinadin NS, Freedman JE, Tanriverdi K, Alsop E, Van Keuren-Jensen K, Sauld JFK, Mahajan G, Khan SS, Colangelo L, Nayor M, Fisher-Hoch S, McCormick JB, North KE, Below JE, Wells QS, Abel ED, Kalhan R, Scott C, Guilliams M, Gamazon ER, Fallowfield JA, Banovich NE, Das S, Shah R. A prognostic molecular signature of hepatic steatosis is spatially heterogeneous and dynamic in human liver. Cell Rep Med 2024; 5:101871. [PMID: 39657669 PMCID: PMC11722105 DOI: 10.1016/j.xcrm.2024.101871] [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: 07/30/2024] [Revised: 09/06/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024]
Abstract
Hepatic steatosis is a central phenotype in multi-system metabolic dysfunction and is increasing in parallel with the obesity pandemic. We use a translational approach integrating clinical phenotyping and outcomes, circulating proteomics, and tissue transcriptomics to identify dynamic, functional biomarkers of hepatic steatosis. Using multi-modality imaging and broad proteomic profiling, we identify proteins implicated in the progression of hepatic steatosis that are largely encoded by genes enriched at the transcriptional level in the human liver. These transcripts are differentially expressed across areas of steatosis in spatial transcriptomics, and several are dynamic during stages of steatosis. Circulating multi-protein signatures of steatosis strongly associate with fatty liver disease and multi-system metabolic outcomes. Using a humanized "liver-on-a-chip" model, we induce hepatic steatosis, confirming cell-specific expression of prioritized targets. These results underscore the utility of this approach to identify a prognostic, functional, dynamic "liquid biopsy" of human liver, relevant to biomarker discovery and mechanistic research applications.
Collapse
Affiliation(s)
- Andrew S Perry
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Niran Hadad
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Emeli Chatterjee
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Maria Jimenez-Ramos
- Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | | | - Rashedeh Roshani
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Michael J Betti
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Shilin Zhao
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Shi Huang
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Liesbet Martens
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Timothy J Kendall
- Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK; Edinburgh Pathology, University of Edinburgh, Edinburgh, UK
| | - Tinne Thone
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | | | - Samuel Bailin
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Curtis L Gabriel
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John Koethe
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - J Jeffrey Carr
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | | - Jane E Freedman
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | - Eric Alsop
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | | | | | | | - Sadiya S Khan
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Laura Colangelo
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Matthew Nayor
- Sections of Cardiovascular Medicine and Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Susan Fisher-Hoch
- School of Public Health, The University of Texas Health Science Center at Houston, Brownsville, TX, USA
| | - Joseph B McCormick
- School of Public Health, The University of Texas Health Science Center at Houston, Brownsville, TX, USA
| | - Kari E North
- CVD Genetic Epidemiology Computational Laboratory, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Jennifer E Below
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Quinn S Wells
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, USA
| | - Ravi Kalhan
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Charlotte Scott
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Martin Guilliams
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Eric R Gamazon
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | | - Saumya Das
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
| | - Ravi Shah
- Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
4
|
Gaertner K, Terzioglu M, Michell C, Tapanainen R, Pohjoismäki J, Dufour E, Saari S. Species differences in glycerol-3-phosphate metabolism reveals trade-offs between metabolic adaptations and cell proliferation. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2024; 1866:149530. [PMID: 39631556 DOI: 10.1016/j.bbabio.2024.149530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/05/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
The temperate climate-adapted brown hare (Lepus europaeus) and the cold-adapted mountain hare (Lepus timidus) are closely related and interfertile species. However, their skin fibroblasts display distinct gene expression profiles related to fundamental cellular processes. This indicates important metabolic divergence between the two species. Through targeted metabolomics and metabolite tracing, we identified species-specific variations in glycerol 3-phosphate (G3P) metabolism. G3P is a key metabolite of the G3P shuttle, which transfers reducing equivalents from cytosolic NADH to the mitochondrial electron transport chain (ETC), consequently regulating glycolysis, lipid metabolism, and mitochondrial bioenergetics. Alterations in G3P metabolism have been implicated in multiple human pathologies including cancer and diabetes. We observed that mountain hare mitochondria exhibit elevated G3P shuttle activity, alongside increased membrane potential and decreased mitochondrial temperature. Silencing mitochondrial G3P dehydrogenase (GPD2), which couples the conversion of G3P to the ETC, uncovered its species-specific role in controlling mitochondrial membrane potential and highlighted its involvement in skin fibroblast thermogenesis. Unexpectedly, GPD2 silencing enhanced wound healing and cell proliferation rates in a species-specific manner. Our study underscores the pivotal role of the G3P shuttle in mediating physiological, bioenergetic, and metabolic divergence between these hare species.
Collapse
Affiliation(s)
- Kateryna Gaertner
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mügen Terzioglu
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Craig Michell
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland; King Abdullah University of Science and Technology, Makkah, Saudi Arabia
| | - Riikka Tapanainen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| | - Jaakko Pohjoismäki
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| | - Eric Dufour
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | - Sina Saari
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| |
Collapse
|
5
|
Gurung S, Karamched S, Perocheau D, Seunarine KK, Baldwin T, Alrashidi H, Touramanidou L, Duff C, Elkhateeb N, Stepien KM, Sharma R, Morris A, Hartley T, Crowther L, Grunewald S, Cleary M, Mundy H, Chakrapani A, Batzios S, Davison J, Footitt E, Tuschl K, Lachmann R, Murphy E, Santra S, Uudelepp M, Yeo M, Finn PF, Cavedon A, Siddiqui S, Rice L, Martini PGV, Frassetto A, Heales S, Mills PB, Gissen P, Clayden JD, Clark CA, Eaton S, Kalber TL, Baruteau J. The incidence of movement disorder increases with age and contrasts with subtle and limited neuroimaging abnormalities in argininosuccinic aciduria. J Inherit Metab Dis 2024; 47:1213-1227. [PMID: 38044746 PMCID: PMC11586606 DOI: 10.1002/jimd.12691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/01/2023] [Accepted: 11/03/2023] [Indexed: 12/05/2023]
Abstract
Argininosuccinate lyase (ASL) is integral to the urea cycle detoxifying neurotoxic ammonia and the nitric oxide (NO) biosynthesis cycle. Inherited ASL deficiency causes argininosuccinic aciduria (ASA), a rare disease with hyperammonemia and NO deficiency. Patients present with developmental delay, epilepsy and movement disorder, associated with NO-mediated downregulation of central catecholamine biosynthesis. A neurodegenerative phenotype has been proposed in ASA. To better characterise this neurodegenerative phenotype in ASA, we conducted a retrospective study in six paediatric and adult metabolic centres in the UK in 2022. We identified 60 patients and specifically looked for neurodegeneration-related symptoms: movement disorder such as ataxia, tremor and dystonia, hypotonia/fatigue and abnormal behaviour. We analysed neuroimaging with diffusion tensor imaging (DTI) magnetic resonance imaging (MRI) in an individual with ASA with movement disorders. We assessed conventional and DTI MRI alongside single photon emission computer tomography (SPECT) with dopamine analogue radionuclide 123I-ioflupane, in Asl-deficient mice treated by hASL mRNA with normalised ureagenesis. Movement disorders in ASA appear in the second and third decades of life, becoming more prevalent with ageing and independent from the age of onset of hyperammonemia. Neuroimaging can show abnormal DTI features affecting both grey and white matter, preferentially basal ganglia. ASA mouse model with normalised ureagenesis did not recapitulate these DTI findings and showed normal 123I-ioflupane SPECT and cerebral dopamine metabolomics. Altogether these findings support the pathophysiology of a late-onset movement disorder with cell-autonomous functional central catecholamine dysregulation but without or limited neurodegeneration of dopaminergic neurons, making these symptoms amenable to targeted therapy.
Collapse
|
6
|
Häberle J, Siri B, Dionisi‐Vici C. Quo vadis ureagenesis disorders? A journey from 90 years ago into the future. J Inherit Metab Dis 2024; 47:1120-1128. [PMID: 38837457 PMCID: PMC11586591 DOI: 10.1002/jimd.12763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/30/2024] [Accepted: 05/14/2024] [Indexed: 06/07/2024]
Abstract
The pathway of ammonia disposal in the mammalian organism has been described in 1932 as a metabolic cycle present in the liver in different compartments. In 1958, the first human disorder affecting this pathway was described as a genetic condition leading to cognitive impairment and constant abnormalities of amino acid metabolism. Since then, defects in all enzymes and transporters of the urea cycle have been described, referring to them as primary urea cycle disorders causing primary hyperammonemia. In addition, there is a still increasing list of conditions that impact on the function of the urea cycle by various mechanisms, hereby leading to secondary hyperammonemia. Despite great advances in understanding the molecular background and the biochemical specificities of both primary and secondary hyperammonemias, there remain many open questions: we do not fully understand the pathophysiology in many of the conditions; we do not always understand the highly variable clinical course of affected patients; we clearly appreciate the need for novel and improved diagnostic and therapeutic approaches. This study does look back to the beginning of the urea cycle (hi)story, briefly describes the journey through past decades, hereby illustrating advancements and knowledge gaps, and gives examples for the extremely broad perspective imminent to some of the defects of ureagenesis and allied conditions.
Collapse
Affiliation(s)
- Johannes Häberle
- Division of Metabolism and Children's Research CenterUniversity Children's Hospital Zurich, University of ZurichZurichSwitzerland
| | - Barbara Siri
- Division of Metabolic Diseases and HepatologyBambino Gesù Children's Hospital IRCCSRomeItaly
| | - Carlo Dionisi‐Vici
- Division of Metabolic Diseases and HepatologyBambino Gesù Children's Hospital IRCCSRomeItaly
| |
Collapse
|
7
|
Ghosh N, Mahalanobish S, Sil PC. Reprogramming of urea cycle in cancer: Mechanism, regulation and prospective therapeutic scopes. Biochem Pharmacol 2024; 228:116326. [PMID: 38815626 DOI: 10.1016/j.bcp.2024.116326] [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: 01/17/2024] [Revised: 05/27/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Hepatic urea cycle, previously known as ornithine cycle, is the chief biochemical pathway that deals with the disposal of excessive nitrogen in form of urea, resulted from protein breakdown and concomitant condensation of ammonia. Enzymes involved in urea cycle are expressed differentially outside hepatic tissue and are mostly involved in production of arginine from citrulline in arginine-depleted condition. Inline, cancer cells frequently adapt metabolic rewiring to support sufficient biomass production in order to sustain tumor cell survival, multiplication and subsequent growth. For the accomplishment of this aim, metabolic reprogramming in cancer cells is set in way so that cellular nitrogen and carbon repertoire can be utilized and channelized maximally towards anabolic reactions. A strategy to meet such outcome is to cut down unnecessary catabolic reactions and nitrogen elimination. Thus, transfigured urea cycle is a hallmark of neoplasia. During oncogenesis, altered expression and regulation of enzymes involved in urea cycle is a revolutionary approach meet to maximum incorporation of nitrogen for sustaining tumor specific biogenesis. Currently, we have reviewed neoplasm-specific deregulations of urea cycle-enzymes in different types and stages of cancers suggesting its context-oriented dynamic nature. Considering such insight to be valuable in terms of prospective cancer diagnosis and therapeutics adaptive evolution of deregulated urea cycle has been enlightened.
Collapse
Affiliation(s)
- Noyel Ghosh
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, West Bengal, India
| | - Sushweta Mahalanobish
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, West Bengal, India
| | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, West Bengal, India.
| |
Collapse
|
8
|
Vucko E, Baker J, Becker K, Havens K, Arduini K, Shim S. Management of a urea cycle disorder in the setting of socioeconomic and language barriers. Mol Genet Metab Rep 2024; 39:101108. [PMID: 39309541 PMCID: PMC11412918 DOI: 10.1016/j.ymgmr.2024.101108] [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: 01/26/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 09/25/2024] Open
Abstract
Argininosuccinic aciduria (ASA) is a disorder that results from a deficiency in the urea cycle enzyme argininosuccinate lyase. Variable manifestations of this hereditary disorder are associated with hyperammonemia and can include lethargy, somnolence, and respiratory alkalosis in neonates, and vomiting, headaches, and neurocognitive deficiencies later in life. Management of ASA includes rapid measures to address hyperammonemia and long-term steps to maintain metabolic stability. Management paradigms should also consider social determinants of health, which are non-medical factors that influence health outcomes. Here, we describe the case of a male pediatric patient with ASA whose treatment has included considerations for his family's refugee status, language barriers, cultural adjustments, limited income, and transportation challenges.
Collapse
Affiliation(s)
- Erika Vucko
- Division of Genetics, Genomics & Metabolism, Ann & Robert H. Lurie Children's Hospital of Chicago, 225 E Chicago Ave, Chicago, IL 60611, USA
- Department of Pediatrics (Genetics, Genomics, and Metabolism), Northwestern University Feinberg School of Medicine, 420 E Superior St, Chicago, IL 60611, USA
| | - Joshua Baker
- Division of Genetics, Genomics & Metabolism, Ann & Robert H. Lurie Children's Hospital of Chicago, 225 E Chicago Ave, Chicago, IL 60611, USA
- Department of Pediatrics (Genetics, Genomics, and Metabolism), Northwestern University Feinberg School of Medicine, 420 E Superior St, Chicago, IL 60611, USA
| | - Karen Becker
- Division of Genetics, Genomics & Metabolism, Ann & Robert H. Lurie Children's Hospital of Chicago, 225 E Chicago Ave, Chicago, IL 60611, USA
| | - Kirsten Havens
- Division of Genetics, Genomics & Metabolism, Ann & Robert H. Lurie Children's Hospital of Chicago, 225 E Chicago Ave, Chicago, IL 60611, USA
| | - Katherine Arduini
- Division of Genetics, Genomics & Metabolism, Ann & Robert H. Lurie Children's Hospital of Chicago, 225 E Chicago Ave, Chicago, IL 60611, USA
| | - Soo Shim
- Division of Genetics, Genomics & Metabolism, Ann & Robert H. Lurie Children's Hospital of Chicago, 225 E Chicago Ave, Chicago, IL 60611, USA
| |
Collapse
|
9
|
Shen G, Liu J, Yang H, Xie N, Yang Y. mRNA therapies: Pioneering a new era in rare genetic disease treatment. J Control Release 2024; 369:696-721. [PMID: 38580137 DOI: 10.1016/j.jconrel.2024.03.056] [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: 11/04/2023] [Revised: 03/16/2024] [Accepted: 03/30/2024] [Indexed: 04/07/2024]
Abstract
Rare genetic diseases, often referred to as orphan diseases due to their low prevalence and limited treatment options, have long posed significant challenges to our medical system. In recent years, Messenger RNA (mRNA) therapy has emerged as a highly promising treatment approach for various diseases caused by genetic mutations. Chemically modified mRNA is introduced into cells using carriers like lipid-based nanoparticles (LNPs), producing functional proteins that compensate for genetic deficiencies. Given the advantages of precise dosing, biocompatibility, transient expression, and minimal risk of genomic integration, mRNA therapies can safely and effectively correct genetic defects in rare diseases and improve symptoms. Currently, dozens of mRNA drugs targeting rare diseases are undergoing clinical trials. This comprehensive review summarizes the progress of mRNA therapy in treating rare genetic diseases. It introduces the development, molecular design, and delivery systems of mRNA therapy, highlighting their research progress in rare genetic diseases based on protein replacement and gene editing. The review also summarizes research progress in various rare disease models and clinical trials. Additionally, it discusses the challenges and future prospects of mRNA therapy. Researchers are encouraged to join this field and collaborate to advance the clinical translation of mRNA therapy, bringing hope to patients with rare genetic diseases.
Collapse
Affiliation(s)
- Guobo Shen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jian Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hanmei Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Na Xie
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China.
| | - Yang Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
10
|
Jalil S, Keskinen T, Juutila J, Sartori Maldonado R, Euro L, Suomalainen A, Lapatto R, Kuuluvainen E, Hietakangas V, Otonkoski T, Hyvönen ME, Wartiovaara K. Genetic and functional correction of argininosuccinate lyase deficiency using CRISPR adenine base editors. Am J Hum Genet 2024; 111:714-728. [PMID: 38579669 PMCID: PMC11023919 DOI: 10.1016/j.ajhg.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 04/07/2024] Open
Abstract
Argininosuccinate lyase deficiency (ASLD) is a recessive metabolic disorder caused by variants in ASL. In an essential step in urea synthesis, ASL breaks down argininosuccinate (ASA), a pathognomonic ASLD biomarker. The severe disease forms lead to hyperammonemia, neurological injury, and even early death. The current treatments are unsatisfactory, involving a strict low-protein diet, arginine supplementation, nitrogen scavenging, and in some cases, liver transplantation. An unmet need exists for improved, efficient therapies. Here, we show the potential of a lipid nanoparticle-mediated CRISPR approach using adenine base editors (ABEs) for ASLD treatment. To model ASLD, we first generated human-induced pluripotent stem cells (hiPSCs) from biopsies of individuals homozygous for the Finnish founder variant (c.1153C>T [p.Arg385Cys]) and edited this variant using the ABE. We then differentiated the hiPSCs into hepatocyte-like cells that showed a 1,000-fold decrease in ASA levels compared to those of isogenic non-edited cells. Lastly, we tested three different FDA-approved lipid nanoparticle formulations to deliver the ABE-encoding RNA and the sgRNA targeting the ASL variant. This approach efficiently edited the ASL variant in fibroblasts with no apparent cell toxicity and minimal off-target effects. Further, the treatment resulted in a significant decrease in ASA, to levels of healthy donors, indicating restoration of the urea cycle. Our work describes a highly efficient approach to editing the disease-causing ASL variant and restoring the function of the urea cycle. This method relies on RNA delivered by lipid nanoparticles, which is compatible with clinical applications, improves its safety profile, and allows for scalable production.
Collapse
Affiliation(s)
- Sami Jalil
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Timo Keskinen
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Juhana Juutila
- Faculty of Biological and Environmental Sciences University of Helsinki, Helsinki, Finland; Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Rocio Sartori Maldonado
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Liliya Euro
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anu Suomalainen
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Risto Lapatto
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Emilia Kuuluvainen
- Faculty of Biological and Environmental Sciences University of Helsinki, Helsinki, Finland; Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Ville Hietakangas
- Faculty of Biological and Environmental Sciences University of Helsinki, Helsinki, Finland; Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Timo Otonkoski
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mervi E Hyvönen
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Kirmo Wartiovaara
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Clinical Genetics, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
11
|
Hajaj E, Pozzi S, Erez A. From the Inside Out: Exposing the Roles of Urea Cycle Enzymes in Tumors and Their Micro and Macro Environments. Cold Spring Harb Perspect Med 2024; 14:a041538. [PMID: 37696657 PMCID: PMC10982720 DOI: 10.1101/cshperspect.a041538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
Catabolic pathways change in anabolic diseases such as cancer to maintain metabolic homeostasis. The liver urea cycle (UC) is the main catabolic pathway for disposing excess nitrogen. Outside the liver, the UC enzymes are differentially expressed based on each tissue's needs for UC intermediates. In tumors, there are changes in the expression of UC enzymes selected for promoting tumorigenesis by increasing the availability of essential UC substrates and products. Consequently, there are compensatory changes in the expression of UC enzymes in the cells that compose the tumor microenvironment. Moreover, extrahepatic tumors induce changes in the expression of the liver UC, which contribute to the systemic manifestations of cancer, such as weight loss. Here, we review the multilayer changes in the expression of UC enzymes throughout carcinogenesis. Understanding the changes in UC expression in the tumor and its micro and macro environment can help identify biomarkers for early cancer diagnosis and vulnerabilities that can be targeted for therapy.
Collapse
Affiliation(s)
- Emma Hajaj
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Sabina Pozzi
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ayelet Erez
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
12
|
Martelli F, Lin J, Mele S, Imlach W, Kanca O, Barlow CK, Paril J, Schittenhelm RB, Christodoulou J, Bellen HJ, Piper MDW, Johnson TK. Identifying potential dietary treatments for inherited metabolic disorders using Drosophila nutrigenomics. Cell Rep 2024; 43:113861. [PMID: 38416643 PMCID: PMC11037929 DOI: 10.1016/j.celrep.2024.113861] [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: 05/16/2023] [Revised: 12/09/2023] [Accepted: 02/08/2024] [Indexed: 03/01/2024] Open
Abstract
Inherited metabolic disorders are a group of genetic conditions that can cause severe neurological impairment and child mortality. Uniquely, these disorders respond to dietary treatment; however, this option remains largely unexplored because of low disorder prevalence and the lack of a suitable paradigm for testing diets. Here, we screened 35 Drosophila amino acid disorder models for disease-diet interactions and found 26 with diet-altered development and/or survival. Using a targeted multi-nutrient array, we examine the interaction in a model of isolated sulfite oxidase deficiency, an infant-lethal disorder. We show that dietary cysteine depletion normalizes their metabolic profile and rescues development, neurophysiology, behavior, and lifelong fly survival, thus providing a basis for further study into the pathogenic mechanisms involved in this disorder. Our work highlights the diet-sensitive nature of metabolic disorders and establishes Drosophila as a valuable tool for nutrigenomic studies for informing potential dietary therapies.
Collapse
Affiliation(s)
- Felipe Martelli
- School of Biological Sciences, Monash University, Clayton, VIC 3800, Australia
| | - Jiayi Lin
- School of Biological Sciences, Monash University, Clayton, VIC 3800, Australia
| | - Sarah Mele
- School of Biological Sciences, Monash University, Clayton, VIC 3800, Australia
| | - Wendy Imlach
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Oguz Kanca
- Department of Molecular and Human Genetics and Duncan Neurological Research Institute at Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Christopher K Barlow
- Monash Proteomics & Metabolomics Facility, Monash Biomedicine Discovery Institute & Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Jefferson Paril
- School of BioSciences, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics & Metabolomics Facility, Monash Biomedicine Discovery Institute & Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - John Christodoulou
- Murdoch Children's Research Institute, Parkville, VIC 3052, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Hugo J Bellen
- Department of Molecular and Human Genetics and Duncan Neurological Research Institute at Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Matthew D W Piper
- School of Biological Sciences, Monash University, Clayton, VIC 3800, Australia.
| | - Travis K Johnson
- School of Biological Sciences, Monash University, Clayton, VIC 3800, Australia; Department of Biochemistry and Chemistry and La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia.
| |
Collapse
|
13
|
Perry AS, Hadad N, Chatterjee E, Ramos MJ, Farber-Eger E, Roshani R, Stolze LK, Zhao S, Martens L, Kendall TJ, Thone T, Amancherla K, Bailin S, Gabriel CL, Koethe J, Carr JJ, Terry JG, Freedman J, Tanriverdi K, Alsop E, Keuren-Jensen KV, Sauld JFK, Mahajan G, Khan S, Colangelo L, Nayor M, Fisher-Hoch S, McCormick J, North KE, Below J, Wells Q, Abel D, Kalhan R, Scott C, Guilliams M, Fallowfield JA, Banovich NE, Das S, Shah R. A prognostic molecular signature of hepatic steatosis is spatially heterogeneous and dynamic in human liver. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.26.24301828. [PMID: 38352394 PMCID: PMC10863022 DOI: 10.1101/2024.01.26.24301828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) prevalence is increasing in parallel with an obesity pandemic, calling for novel strategies for prevention and treatment. We defined a circulating proteome of human MASLD across ≈7000 proteins in ≈5000 individuals from diverse, at-risk populations across the metabolic health spectrum, demonstrating reproducible diagnostic performance and specifying both known and novel metabolic pathways relevant to MASLD (central carbon and amino acid metabolism, hepatocyte regeneration, inflammation, fibrosis, insulin sensitivity). A parsimonious proteomic signature of MASLD was associated with a protection from MASLD and its related multi-system metabolic consequences in >26000 free-living individuals, with an additive effect to polygenic risk. The MASLD proteome was encoded by genes that demonstrated transcriptional enrichment in liver, with spatial transcriptional activity in areas of steatosis in human liver biopsy and dynamicity for select targets in human liver across stages of steatosis. We replicated several top relations from proteomics and spatial tissue transcriptomics in a humanized "liver-on-a-chip" model of MASLD, highlighting the power of a full translational approach to discovery in MASLD. Collectively, these results underscore utility of blood-based proteomics as a dynamic "liquid biopsy" of human liver relevant to clinical biomarker and mechanistic applications.
Collapse
|
14
|
Gurung S, Timmermand OV, Perocheau D, Gil-Martinez AL, Minnion M, Touramanidou L, Fang S, Messina M, Khalil Y, Spiewak J, Barber AR, Edwards RS, Pinto PL, Finn PF, Cavedon A, Siddiqui S, Rice L, Martini PGV, Ridout D, Heywood W, Hargreaves I, Heales S, Mills PB, Waddington SN, Gissen P, Eaton S, Ryten M, Feelisch M, Frassetto A, Witney TH, Baruteau J. mRNA therapy corrects defective glutathione metabolism and restores ureagenesis in preclinical argininosuccinic aciduria. Sci Transl Med 2024; 16:eadh1334. [PMID: 38198573 PMCID: PMC7615535 DOI: 10.1126/scitranslmed.adh1334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 10/06/2023] [Indexed: 01/12/2024]
Abstract
The urea cycle enzyme argininosuccinate lyase (ASL) enables the clearance of neurotoxic ammonia and the biosynthesis of arginine. Patients with ASL deficiency present with argininosuccinic aciduria, an inherited metabolic disease with hyperammonemia and a systemic phenotype coinciding with neurocognitive impairment and chronic liver disease. Here, we describe the dysregulation of glutathione biosynthesis and upstream cysteine utilization in ASL-deficient patients and mice using targeted metabolomics and in vivo positron emission tomography (PET) imaging using (S)-4-(3-18F-fluoropropyl)-l-glutamate ([18F]FSPG). Up-regulation of cysteine metabolism contrasted with glutathione depletion and down-regulated antioxidant pathways. To assess hepatic glutathione dysregulation and liver disease, we present [18F]FSPG PET as a noninvasive diagnostic tool to monitor therapeutic response in argininosuccinic aciduria. Human hASL mRNA encapsulated in lipid nanoparticles improved glutathione metabolism and chronic liver disease. In addition, hASL mRNA therapy corrected and rescued the neonatal and adult Asl-deficient mouse phenotypes, respectively, enhancing ureagenesis. These findings provide mechanistic insights in liver glutathione metabolism and support clinical translation of mRNA therapy for argininosuccinic aciduria.
Collapse
Affiliation(s)
- Sonam Gurung
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | | | - Dany Perocheau
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Ana Luisa Gil-Martinez
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Magdalena Minnion
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
- Southampton NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Loukia Touramanidou
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Sherry Fang
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Martina Messina
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Youssef Khalil
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Justyna Spiewak
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Abigail R Barber
- School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Richard S Edwards
- School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Patricia Lipari Pinto
- Santa Maria's Hospital, Lisbon North University Hospital Center, 1649-028 Lisbon, Portugal
| | | | | | | | - Lisa Rice
- Moderna Inc., Cambridge, MA 02139, USA
| | | | - Deborah Ridout
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Wendy Heywood
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Ian Hargreaves
- Pharmacy and Biomolecular Sciences, Liverpool John Moore University, Liverpool L3 5UG, UK
| | - Simon Heales
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Philippa B Mills
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Simon N Waddington
- EGA Institute for Women's Health, University College London, London WC1E 6HX, UK
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of Witswatersrand, Braamfontein, 2000 Johannesburg, South Africa
| | - Paul Gissen
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
- National Institute of Health Research Great Ormond Street Biomedical Research Centre, London WC1N 1EH, UK
| | - Simon Eaton
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Mina Ryten
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
- Southampton NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | | | - Timothy H Witney
- School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Julien Baruteau
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
- National Institute of Health Research Great Ormond Street Biomedical Research Centre, London WC1N 1EH, UK
| |
Collapse
|
15
|
Heng TYJ, Ow JR, Koh AL, Lim JSC, Ong CBK, Goh JCY, Lim JY, Chiou FK, Jamuar SS. To B(enign) or Not to B: functionalisation of variant in a mild form of argininosuccinate lyase deficiency identified through newborn screening. Clin Dysmorphol 2024; 33:43-49. [PMID: 37865865 DOI: 10.1097/mcd.0000000000000475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2023]
Abstract
Argininosuccinate lyase (ASL) deficiency is an autosomal recessive disorder of the urea cycle with a diverse spectrum of clinical presentation that is detectable in newborn screening. We report an 8-year-old girl with ASL deficiency who was detected through newborn screening and was confirmed using biochemical and functional assay. She is compound heterozygous for a likely pathogenic variant NM_000048.4(ASL):c.283C>T (p.Arg95Cys) and a likely benign variant NM_000048.4(ASL): c.1319T>C (p.Leu440Pro). Functional characterisation of the likely benign genetic variant in ASL was performed. Genomic sequencing was performed on the index patient presenting with non-specific symptoms of poor feeding and lethargy and shown to have increased serum and urine argininosuccinic acid. Functional assay using HEK293T cell model was performed. ASL enzymatic activity was reduced for Leu440Pro. This study highlights the role of functional testing of a variant that may appear benign in a patient with a phenotype consistent with ASL deficiency, and reclassifies NM_000048.4(ASL): c.1319T>C (p.Leu440Pro) variant as likely pathogenic.
Collapse
Affiliation(s)
| | - Jin Rong Ow
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR)
| | - Ai Ling Koh
- Genetics Service, Department of Paediatrics, KK Women's and Children's Hospital
- SingHealth Duke-NUS Paediatric Academic Clinical Programme, Duke-NUS Medical School
| | - James Soon Chuan Lim
- Biochemical Genetics and National Expanded Newborn Screening, Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital
| | | | - Jasmine Chew Yin Goh
- Division of Nursing - Nursing Clinical Services, KK Women's and Children's Hospital
| | - Jiin Ying Lim
- Genetics Service, Department of Paediatrics, KK Women's and Children's Hospital
| | - Fang Kuan Chiou
- SingHealth Duke-NUS Paediatric Academic Clinical Programme, Duke-NUS Medical School
- Gastroenterology, Hepatology & Nutrition Service, Department of Paediatrics, KK Women's and Children's Hospital
| | - Saumya Shekhar Jamuar
- Genetics Service, Department of Paediatrics, KK Women's and Children's Hospital
- SingHealth Duke-NUS Paediatric Academic Clinical Programme, Duke-NUS Medical School
- SingHealth Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Republic of Singapore
| |
Collapse
|
16
|
Öztürk F, Ünlü CI, Sarıcaoğlu H, Aydoğan K, Erdöl Ş. Acrodermatitis dysmetabolica with argininosuccinate lyase deficiency. Indian J Dermatol Venereol Leprol 2023; 89:927. [PMID: 37317759 DOI: 10.25259/ijdvl_75_2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/11/2023] [Indexed: 06/16/2023]
Affiliation(s)
- Ferdi Öztürk
- Department of Dermatology, Division of Metabolism, Bursa Uludag University Faculty of Medicine, Bursa, Nilüfer, Bursa, Turkey
| | - Cansu Irmak Ünlü
- Department of Dermatology, Division of Metabolism, Bursa Uludag University Faculty of Medicine, Bursa, Nilüfer, Bursa, Turkey
| | - Hayriye Sarıcaoğlu
- Department of Dermatology, Division of Metabolism, Bursa Uludag University Faculty of Medicine, Bursa, Nilüfer, Bursa, Turkey
| | - Kenan Aydoğan
- Department of Dermatology, Division of Metabolism, Bursa Uludag University Faculty of Medicine, Bursa, Nilüfer, Bursa, Turkey
| | - Şahin Erdöl
- Department of Pediatrics, Division of Metabolism, Bursa Uludag University Faculty of Medicine, Bursa, Nilüfer, Bursa, Turkey
| |
Collapse
|
17
|
Nagayoshi Y, Nakayama M, Nagano H, Morikawa K, Nishi M, Nishihara T, Sakaino N, Kawano H, Tsujita K, Mitsubuchi H. Coronary Vasospasm in a Patient With Argininosuccinic Aciduria. Am J Cardiol 2023; 192:155-159. [PMID: 36807131 DOI: 10.1016/j.amjcard.2023.01.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/14/2023] [Accepted: 01/16/2023] [Indexed: 02/21/2023]
Abstract
A 39-year-old male was referred for treatment of hypertension. He had been treated for argininosuccinic aciduria since 8 months of age. Therapeutic drugs, including l-arginine, sodium phenylbutyrate, and antiepileptic drugs, had been prescribed. A detailed medical history revealed that he complained of chest discomfort under psychologic stress. A 12-lead electrocardiogram showed abnormal q waves in lead III and aVF. Transthoracic echocardiography showed hypokinesia of the left ventricular posterior wall. The patient was diagnosed with myocardial infarction because of coronary vasospastic angina by intracoronary acetylcholine provocation test. Argininosuccinic aciduria is a genetic disorder of the urea cycle caused by a deficiency of argininosuccinate lyase. Reduction of the enzymatic activity leads to a decrease in nitric oxide production, even if arginine is supplemented. Our case report supports the significance of endothelial function in the pathogenesis of coronary vasospasm.
Collapse
Affiliation(s)
- Yasuhiro Nagayoshi
- Department of Cardiology, Amakusa Medical Center, Amakusa City, Japan; Department of Cardiovascular Medicine, Graduate School of Medical Science, Kumamoto University, Kumamoto City, Japan.
| | | | - Haruka Nagano
- Department of Cardiology, Amakusa Medical Center, Amakusa City, Japan
| | - Kei Morikawa
- Department of Cardiology, Amakusa Medical Center, Amakusa City, Japan
| | - Masato Nishi
- Department of Cardiology, Amakusa Medical Center, Amakusa City, Japan
| | - Taiki Nishihara
- Department of Cardiology, Amakusa Medical Center, Amakusa City, Japan
| | - Naritsugu Sakaino
- Department of Cardiology, Amakusa Medical Center, Amakusa City, Japan
| | - Hiroaki Kawano
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kumamoto University, Kumamoto City, Japan
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kumamoto University, Kumamoto City, Japan
| | - Hiroshi Mitsubuchi
- Department of Pediatrics, Graduate School of Medical Science, Kumamoto University, Kumamoto City, Japan; Division of Neonatology, Kumamoto University Hospital, Kumamoto City, Japan
| |
Collapse
|
18
|
Duff C, Baruteau J. Modelling urea cycle disorders using iPSCs. NPJ Regen Med 2022; 7:56. [PMID: 36163209 PMCID: PMC9513077 DOI: 10.1038/s41536-022-00252-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 08/10/2022] [Indexed: 11/23/2022] Open
Abstract
The urea cycle is a liver-based pathway enabling disposal of nitrogen waste. Urea cycle disorders (UCDs) are inherited metabolic diseases caused by deficiency of enzymes or transporters involved in the urea cycle and have a prevalence of 1:35,000 live births. Patients present recurrent acute hyperammonaemia, which causes high rate of death and neurological sequelae. Long-term therapy relies on a protein-restricted diet and ammonia scavenger drugs. Currently, liver transplantation is the only cure. Hence, high unmet needs require the identification of effective methods to model these diseases to generate innovative therapeutics. Advances in both induced pluripotent stem cells (iPSCs) and genome editing technologies have provided an invaluable opportunity to model patient-specific phenotypes in vitro by creating patients' avatar models, to investigate the pathophysiology, uncover novel therapeutic targets and provide a platform for drug discovery. This review summarises the progress made thus far in generating 2- and 3-dimensional iPSCs models for UCDs, the challenges encountered and how iPSCs offer future avenues for innovation in developing the next-generation of therapies for UCDs.
Collapse
Affiliation(s)
- Claire Duff
- Genetics and Genomic Medicine Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Julien Baruteau
- Genetics and Genomic Medicine Department, Great Ormond Street Institute of Child Health, University College London, London, UK.
- National Institute of Health Research Great Ormond Street Biomedical Research Centre, London, UK.
- Metabolic Medicine Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.
| |
Collapse
|
19
|
Fátima Lopes F, Sitta A, de Moura Coelho D, Schmitt Ribas G, Lamberty Faverzani J, Gomes Dos Reis B, Wajner M, Vargas CR. Clinical findings of patients with hyperammonemia affected by urea cycle disorders with hepatic encephalopathy. Int J Dev Neurosci 2022; 82:772-788. [PMID: 36129623 DOI: 10.1002/jdn.10229] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/21/2022] [Accepted: 09/15/2022] [Indexed: 11/10/2022] Open
Abstract
Urea Cycle Disorders (UCD) are a group of genetic diseases caused by deficiencies in the enzymes and transporters involved in the urea cycle. The impairment of the cycle results in ammonia accumulation, leading to neurological dysfunctions and poor outcomes to affected patients. The aim of this study is to investigate and describe UCD patients principal clinical and biochemical presentations to support professionals on urgent diagnosis and quick management, aiming better outcomes for patients. We explored medical records of thirty patients diagnosed in a referral center from Brazil to delineate UCD clinical and biochemical profile. Patients demonstrated a range of signs and symptoms, such as altered levels of consciousness, acute encephalopathy, seizures, progressive loss of appetite, vomiting, coma, and respiratory distress, in most cases combined with high levels of ammonia, which is an immediate biomarker, leading to an UCD suspicion. The most prevalent UCD detected were ornithine transcarbamylase deficiency (11), followed by citrullinemia type I (10), hyperargininemia (5), carbamoyl phosphate synthase 1 deficiency (2) and argininosuccinic aciduria (2). Clinical symptoms were highly severe, being the majority developmental and neurological disabilities, with 20% of death rate. Laboratory analysis revealed high levels of ammonia (mean ± SD: 860 ± 470 μmol/L; reference value: ≤ 80 μmol/L), hypoglycemia, metabolic acidosis, and high excretion of orotic acid in the urine (except in CPS1 deficiency). We emphasize the need of urgent identification of UCD clinical and biochemical conditions, and immediate measurement of ammonia, to enable the correct diagnosis and increase the chances of patients survival, minimizing neurological and psychomotor damage caused by hepatic encephalopathy.
Collapse
Affiliation(s)
- Franciele Fátima Lopes
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Angela Sitta
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | | | | | - Jéssica Lamberty Faverzani
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Bianca Gomes Dos Reis
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Moacir Wajner
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Carmen Regla Vargas
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
20
|
Huang L, He X, Peng W, He X, Xu B, Xu H, Wang Y, Xu W, Chen W, Wang S, Zhou L, Liu N, Xu Y, Lu W. Hyperuricemia induces liver injury by upregulating HIF-1α and inhibiting arginine biosynthesis pathway in mouse liver and human L02 hepatocytes. Biochem Biophys Res Commun 2022; 617:55-61. [PMID: 35696777 DOI: 10.1016/j.bbrc.2022.05.096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/29/2022] [Accepted: 05/30/2022] [Indexed: 11/25/2022]
Abstract
The molecular mechanisms of uric acid (UA)-induced liver injury has not been clearly elucidated. In this study, we aimed to investigate the effect and action mechanisms of UA in liver injury. We analyzed the damaging effect of UA on mouse liver and L02 cells and subsequently performed metabolomics studies on L02 cells to identify abnormal metabolic pathways. Finally, we verified transcription factors that regulate related metabolic enzymes. UA directly activated the hepatic NLRP3 inflammasome and Bax apoptosis pathway invivo and invitro. Related metabolites in the arginine biosynthesis pathway (or urea cycle), l-arginine and l-argininosuccinate were decreased, and ammonia was increased in UA-stimulated L02 cells, which was mediated by carbamoyl phosphate synthase 1 (CPS1), argininosuccinate synthase (ASS) and argininosuccinate lyase (ASL) downregulation. UA upregulated hypoxia inducible factor-1alpha (HIF-1α) invivo and invitro, and HIF-1α inhibition alleviated the UA-induced ASS downregulation and hepatocyte injury. In conclusion, UA upregulates HIF-1α and inhibits urea cycle enzymes (UCEs). This leads to liver injury, with evidence of hepatocyte inflammation, apoptosis and oxidative stress.
Collapse
Affiliation(s)
- Lei Huang
- Basic Medical College, Anhui Medical University, Hefei, 230032, China
| | - Xinyu He
- Basic Medical College, Anhui Medical University, Hefei, 230032, China
| | - Wen Peng
- Department of Oncology, The People's Hospital of Guizhou Province, Guiyang, 550004, China
| | - Xueqing He
- Basic Medical College, Anhui Medical University, Hefei, 230032, China
| | - Bei Xu
- Basic Medical College, Anhui Medical University, Hefei, 230032, China
| | - Hu Xu
- Basic Medical College, Anhui Medical University, Hefei, 230032, China
| | - Yaoxing Wang
- Basic Medical College, Anhui Medical University, Hefei, 230032, China
| | - Wenjun Xu
- Basic Medical College, Anhui Medical University, Hefei, 230032, China
| | - Wentong Chen
- Basic Medical College, Anhui Medical University, Hefei, 230032, China
| | - Sheng Wang
- Center for Scientific Rrsearch, Anhui Medical University, Hefei, 230032, China
| | - Lanlan Zhou
- School of Medical Technology and Nursing, Shenzhen Polytechnic, Shenzhen, 518055, China
| | - Ning Liu
- Basic Medical College, Anhui Medical University, Hefei, 230032, China.
| | - Youzhi Xu
- Basic Medical College, Anhui Medical University, Hefei, 230032, China.
| | - Wenjie Lu
- Basic Medical College, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
21
|
Cardiac disruption of SDHAF4-mediated mitochondrial complex II assembly promotes dilated cardiomyopathy. Nat Commun 2022; 13:3947. [PMID: 35803927 PMCID: PMC9270418 DOI: 10.1038/s41467-022-31548-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/15/2022] [Indexed: 12/30/2022] Open
Abstract
Succinate dehydrogenase, which is known as mitochondrial complex II, has proven to be a fascinating machinery, attracting renewed and increased interest in its involvement in human diseases. Herein, we find that succinate dehydrogenase assembly factor 4 (SDHAF4) is downregulated in cardiac muscle in response to pathological stresses and in diseased hearts from human patients. Cardiac loss of Sdhaf4 suppresses complex II assembly and results in subunit degradation and complex II deficiency in fetal mice. These defects are exacerbated in young adults with globally impaired metabolic capacity and activation of dynamin-related protein 1, which induces excess mitochondrial fission and mitophagy, thereby causing progressive dilated cardiomyopathy and lethal heart failure in animals. Targeting mitochondria via supplementation with fumarate or inhibiting mitochondrial fission improves mitochondrial dynamics, partially restores cardiac function and prolongs the lifespan of mutant mice. Moreover, the addition of fumarate is found to dramatically improve cardiac function in myocardial infarction mice. These findings reveal a vital role for complex II assembly in the development of dilated cardiomyopathy and provide additional insights into therapeutic interventions for heart diseases. Functional succinate dehydrogenase (SDH) complex is vital to mitochondrial homeostasis. Here the authors show that disruption of SDH assembly in the heart causes dilated cardiomyopathy via impairing the mitochondrial integrity and metabolism and that mitochondrial interventions can be an effective approach to ameliorate the disease progression.
Collapse
|
22
|
Kian N, Samieefar N, Rezaei N. Prenatal risk factors and genetic causes of ADHD in children. World J Pediatr 2022; 18:308-319. [PMID: 35235183 DOI: 10.1007/s12519-022-00524-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 02/07/2022] [Indexed: 12/18/2022]
Abstract
BACKGROUND Attention deficit/hyperactivity disorder (ADHD) is a common disease among children; it affected 5-7% of the population in 2015. ADHD is a multifactorial disease, and its etiology is still not clearly understood. DATA SOURCES This narrative review has been done by searching the PubMed and Embase databases using attention deficit/hyperactivity disorder, ADHD, risk factors; genetics; pediatrics; psychiatrics as keywords. RESULTS ADHD is considered to be a hereditary disorder in which genes play the fundamental role in the pathogenesis; however, findings from genetic-environmental studies support the hypothesis that genetic factors can exert effects on an individual's condition by determining his/her responses to environmental exposures, especially those during the prenatal stage. CONCLUSION ADHD is considered as a hereditary disorder in which genes and prenatal risk factors play fundamental roles in the pathogenesis.
Collapse
Affiliation(s)
- Naghmeh Kian
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,USERN Office, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Noosha Samieefar
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,USERN Office, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran. .,Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
Kozłowska L, Santonen T, Duca RC, Godderis L, Jagiello K, Janasik B, Van Nieuwenhuyse A, Poels K, Puzyn T, Scheepers PTJ, Sijko M, Silva MJ, Sosnowska A, Viegas S, Verdonck J, Wąsowicz W. HBM4EU Chromates Study: Urinary Metabolomics Study of Workers Exposed to Hexavalent Chromium. Metabolites 2022; 12:362. [PMID: 35448548 PMCID: PMC9032989 DOI: 10.3390/metabo12040362] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/01/2022] [Accepted: 04/11/2022] [Indexed: 11/30/2022] Open
Abstract
Exposure to hexavalent chromium Cr(VI) may occur in several occupational activities, placing workers in many industries at risk for potential related health outcomes. Untargeted metabolomics was applied to investigate changes in metabolic pathways in response to Cr(VI) exposure. We obtained our data from a study population of 220 male workers with exposure to Cr(VI) and 102 male controls from Belgium, Finland, Poland, Portugal and the Netherlands within the HBM4EU Chromates Study. Urinary metabolite profiles were determined using liquid chromatography mass spectrometry, and differences between post-shift exposed workers and controls were analyzed using principal component analysis. Based on the first two principal components, we observed clustering by industrial chromate application, such as welding, chrome plating, and surface treatment, distinct from controls and not explained by smoking status or alcohol use. The changes in the abundancy of excreted metabolites observed in workers reflect fatty acid and monoamine neurotransmitter metabolism, oxidative modifications of amino acid residues, the excessive formation of abnormal amino acid metabolites and changes in steroid and thyrotropin-releasing hormones. The observed responses could also have resulted from work-related factors other than Cr(VI). Further targeted metabolomics studies are needed to better understand the observed modifications and further explore the suitability of urinary metabolites as early indicators of adverse effects associated with exposure to Cr(VI).
Collapse
Affiliation(s)
- Lucyna Kozłowska
- Laboratory of Human Metabolism Research, Department of Dietetics, Warsaw University of Life Sciences, 02776 Warsaw, Poland;
| | - Tiina Santonen
- Finnish Institute of Occupational Health, 00250 Helsinki, Finland;
| | - Radu Corneliu Duca
- Labotoire National de Santé (LNS), Unit Environmental Hygiene and Human Biological Monitoring, Department of Health Protection, 3555 Dudelange, Luxembourg;
- Centre for Environment and Health, Department of Public Health and Primary Care, KU Leuven (University of Leuven), 3000 Leuven, Belgium; (L.G.); (A.V.N.); (K.P.); (J.V.)
| | - Lode Godderis
- Centre for Environment and Health, Department of Public Health and Primary Care, KU Leuven (University of Leuven), 3000 Leuven, Belgium; (L.G.); (A.V.N.); (K.P.); (J.V.)
- IDEWE, External Service for Prevention and Protection at Work, 3001 Heverlee, Belgium
| | - Karolina Jagiello
- QSAR Laboratory Ltd., 80172 Gdansk, Poland; (K.J.); (T.P.); (A.S.)
- Laboratory of Environmental Chemoinfomatics, Department of Environmental Chemistry and Radiochemistry, Faculty of Chemistry, University of Gdansk, 80308 Gdansk, Poland
| | - Beata Janasik
- Department of Environmental and Biological Monitoring, Nofer Institute of Occupational Medicine, 91348 Lodz, Poland; (B.J.); (W.W.)
| | - An Van Nieuwenhuyse
- Centre for Environment and Health, Department of Public Health and Primary Care, KU Leuven (University of Leuven), 3000 Leuven, Belgium; (L.G.); (A.V.N.); (K.P.); (J.V.)
- Laboratoire National de Santé (LNS), Department of Health Protection, 3555 Dudelange, Luxembourg
| | - Katrien Poels
- Centre for Environment and Health, Department of Public Health and Primary Care, KU Leuven (University of Leuven), 3000 Leuven, Belgium; (L.G.); (A.V.N.); (K.P.); (J.V.)
| | - Tomasz Puzyn
- QSAR Laboratory Ltd., 80172 Gdansk, Poland; (K.J.); (T.P.); (A.S.)
- Laboratory of Environmental Chemoinfomatics, Department of Environmental Chemistry and Radiochemistry, Faculty of Chemistry, University of Gdansk, 80308 Gdansk, Poland
| | - Paul T. J. Scheepers
- Radboud Institute for Health Sciences, Radboudumc, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands;
| | - Monika Sijko
- Laboratory of Human Metabolism Research, Department of Dietetics, Warsaw University of Life Sciences, 02776 Warsaw, Poland;
| | - Maria João Silva
- Human Genetics Department, National Institute of Health Dr. Ricardo Jorge (INSA), Toxicogenomics and Human Health (ToxOmics), NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal;
| | - Anita Sosnowska
- QSAR Laboratory Ltd., 80172 Gdansk, Poland; (K.J.); (T.P.); (A.S.)
| | - Susana Viegas
- Public Health Research Centre, NOVA National School of Public Health, Universidade NOVA de Lisbon, 1600-560 Lisbon, Portugal;
- Comprehensive Health Research Center (CHRC), 1169-056 Lisbon, Portugal
| | - Jelle Verdonck
- Centre for Environment and Health, Department of Public Health and Primary Care, KU Leuven (University of Leuven), 3000 Leuven, Belgium; (L.G.); (A.V.N.); (K.P.); (J.V.)
| | - Wojciech Wąsowicz
- Department of Environmental and Biological Monitoring, Nofer Institute of Occupational Medicine, 91348 Lodz, Poland; (B.J.); (W.W.)
| | | | | |
Collapse
|
24
|
Khare S, Kim LC, Lobel G, Doulias PT, Ischiropoulos H, Nissim I, Keith B, Simon MC. ASS1 and ASL suppress growth in clear cell renal cell carcinoma via altered nitrogen metabolism. Cancer Metab 2021; 9:40. [PMID: 34861885 PMCID: PMC8642968 DOI: 10.1186/s40170-021-00271-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/08/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Kidney cancer is a common adult malignancy in the USA. Clear cell renal cell carcinoma (ccRCC), the predominant subtype of kidney cancer, is characterized by widespread metabolic changes. Urea metabolism is one such altered pathway in ccRCC. The aim of this study was to elucidate the contributions of urea cycle enzymes, argininosuccinate synthase 1 (ASS1), and argininosuccinate lyase (ASL) towards ccRCC progression. METHODS We employed a combination of computational, genetic, and metabolomic tools along with in vivo animal models to establish a tumor-suppressive role for ASS1 and ASL in ccRCC. RESULTS We show that the mRNA and protein expression of urea cycle enzymes ASS1 and ASL are reduced in ccRCC tumors when compared to the normal kidney. Furthermore, the loss of ASL in HK-2 cells (immortalized renal epithelial cells) promotes growth in 2D and 3D growth assays, while combined re-expression of ASS1 and ASL in ccRCC cell lines suppresses growth in 2D, 3D, and in vivo xenograft models. We establish that this suppression is dependent on their enzymatic activity. Finally, we demonstrate that conservation of cellular aspartate, regulation of nitric oxide synthesis, and pyrimidine production play pivotal roles in ASS1+ASL-mediated growth suppression in ccRCC. CONCLUSIONS ccRCC tumors downregulate the components of the urea cycle including the enzymes argininosuccinate synthase 1 (ASS1) and argininosuccinate lyase (ASL). These cytosolic enzymes lie at a critical metabolic hub in the cell and are involved in aspartate catabolism and arginine and nitric oxide biosynthesis. Loss of ASS1 and ASL helps cells redirect aspartate towards pyrimidine synthesis and support enhanced proliferation. Additionally, reduced levels of ASS1 and ASL might help regulate nitric oxide (NO) generation and mitigate its cytotoxic effects. Overall, our work adds to the understanding of urea cycle enzymes in a context-independent of ureagenesis, their role in ccRCC progression, and uncovers novel potential metabolic vulnerabilities in ccRCC.
Collapse
Affiliation(s)
- Sanika Khare
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Laura C Kim
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Graham Lobel
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Paschalis-Thomas Doulias
- Children's Hospital of Philadelphia Research Institute and Departments of Pediatrics and Pharmacology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Harry Ischiropoulos
- Children's Hospital of Philadelphia Research Institute and Departments of Pediatrics and Pharmacology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Itzhak Nissim
- Division of Genetics and Metabolism, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pediatrics, Biochemistry, and Biophysics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Brian Keith
- The Wistar Institute, Philadelphia, PA, 19104, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
25
|
Inherited metabolic diseases: aminoacidopathies, organic acidemia, defects of mitochondrial β-oxidation. A brief overview. ACTA BIOMEDICA SCIENTIFICA 2021. [DOI: 10.29413/abs.2021-6.5.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Inherited metabolic diseases are a large group of inherited monogenic diseases. Metabolic disorders can cause child disability and mortality. Tandem mass spectrometry is a powerful technology that allows to diagnosis a large number of hereditary metabolic diseases. Clinical manifestations are variable, but more often the damages of nervous system, heart, liver, kidneys, hyperammonemia, hypo/hyperglycemia take place. The disease can make its debut at any age, but the severe forms of the disease manifest at infancy. Early diagnosis and treatment can significantly improve the prognosis; many countries expand the list of diseases included in screening programs. At the beginning of 2021 in most regions of the Russian Federation mass newborn screening is carried out for five hereditary metabolic diseases. The age and the range of clinical manifestation are variable; therefore, knowledge of this pathology is very important both for pediatricians and therapists, and for specialized doctors. The article presents a brief description of next groups of metabolic diseases: aminoacidopathies, organic acidurias and fatty acid oxidation defects.
Collapse
|
26
|
Leuger L, Dieu X, Chao de la Barca JM, Moriconi M, Halley G, Donin de Rosière X, Reynier P, Mirebeau‐Prunier D, Homedan C. Late-onset argininosuccinic aciduria in a 72-year-old man presenting with fatal hyperammonemia. JIMD Rep 2021; 62:44-48. [PMID: 34765397 PMCID: PMC8574183 DOI: 10.1002/jmd2.12251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 11/07/2022] Open
Abstract
Argininosuccinate lyase deficiency (ASLD, MIM #207900) is an inherited urea cycle disorder. There are mainly two clinical forms, an acute neonatal form which manifests as life-threatening hyperammonemia, and a late-onset form characterised by polymorphic neuro-cognitive or psychiatric presentation with transient hyperammonemia episodes. Here, we report a late-onset case of ASLD in a 72-year-old man carrying a homozygous pathogenic variant in the exon 16 of the ASL gene, presenting for the first time with fatal hyperammonemic coma. This case report shows the need to systematically carry out an ammonia assay when faced with an unexplained coma.
Collapse
Affiliation(s)
- Laurent Leuger
- Laboratoire de Biochimie et biologie moléculaire, Centre Hospitalier Universitaire d'AngersAngers Cedex 9France
| | - Xavier Dieu
- Laboratoire de Biochimie et biologie moléculaire, Centre Hospitalier Universitaire d'AngersAngers Cedex 9France
| | | | - Mikael Moriconi
- Service de Réanimation Polyvalente et Unité de soins continus, Centre Hospitalier de CornouailleQuimper CedexFrance
| | - Guillaume Halley
- Service de Réanimation Polyvalente et Unité de soins continus, Centre Hospitalier de CornouailleQuimper CedexFrance
| | | | - Pascal Reynier
- Laboratoire de Biochimie et biologie moléculaire, Centre Hospitalier Universitaire d'AngersAngers Cedex 9France
| | - Delphine Mirebeau‐Prunier
- Laboratoire de Biochimie et biologie moléculaire, Centre Hospitalier Universitaire d'AngersAngers Cedex 9France
| | - Chadi Homedan
- Laboratoire de Biochimie et biologie moléculaire, Centre Hospitalier Universitaire d'AngersAngers Cedex 9France
| |
Collapse
|
27
|
Hajaj E, Sciacovelli M, Frezza C, Erez A. The context-specific roles of urea cycle enzymes in tumorigenesis. Mol Cell 2021; 81:3749-3759. [PMID: 34469752 DOI: 10.1016/j.molcel.2021.08.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/13/2021] [Accepted: 07/30/2021] [Indexed: 12/11/2022]
Abstract
The expression of the urea cycle (UC) proteins is dysregulated in multiple cancers, providing metabolic benefits to tumor survival, proliferation, and growth. Here, we review the main changes described in the expression of UC enzymes and metabolites in different cancers at various stages and suggest that these changes are dynamic and should hence be viewed in a context-specific manner. Understanding the evolvability in the activity of the UC pathway in cancer has implications for cancer-immune cell interactions and for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Emma Hajaj
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Marco Sciacovelli
- Medical Research Council Cancer Unit, University of Cambridge, Box 197, Biomedical Campus, Cambridge CB2 0XZ, UK
| | - Christian Frezza
- Medical Research Council Cancer Unit, University of Cambridge, Box 197, Biomedical Campus, Cambridge CB2 0XZ, UK.
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
28
|
Kido J, Matsumoto S, Häberle J, Nakajima Y, Wada Y, Mochizuki N, Murayama K, Lee T, Mochizuki H, Watanabe Y, Horikawa R, Kasahara M, Nakamura K. Long-term outcome of urea cycle disorders: Report from a nationwide study in Japan. J Inherit Metab Dis 2021; 44:826-837. [PMID: 33840128 DOI: 10.1002/jimd.12384] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 12/20/2022]
Abstract
Urea cycle disorders (UCDs) are inherited metabolic disorders with impaired nitrogen detoxification caused by defects in urea cycle enzymes. They often manifest with hyperammonemic attacks resulting in significant morbidity or death. We performed a nationwide questionnaire-based study between January 2000 and March 2018 to document all UCDs in Japan, including diagnoses, treatments, and outcomes. A total of 229 patients with UCDs were enrolled in this study: 73 males and 53 females with ornithine transcarbamylase deficiency (OTCD), 33 patients with carbamoylphosphate synthetase 1 deficiency, 48 with argininosuccinate synthetase deficiency, 14 with argininosuccinate lyase deficiency, and 8 with arginase deficiency. Survival rates at 20 years of age of male and female patients with late-onset OTCD were 100% and 97.7%, respectively. Blood ammonia levels and time of onset had a significant impact on the neurodevelopmental outcome (P < .001 and P = .028, respectively). Hemodialysis and liver transplantation did not prevent poor neurodevelopmental outcomes. While treatment including medication, hemodialysis, and liver transplantation may aid in decreasing blood ammonia and/or preventing severe hyperammonemia, a blood ammonia level ≥ 360 μmol/L was found to be a significant indicator for a poor neurodevelopmental outcome. In conclusion, although current therapy for UCDs has advanced and helped saving lives, patients with blood ammonia levels ≥ 360 μmol/L at onset often have impaired neurodevelopmental outcomes. Novel neuroprotective measures should therefore be developed to achieve better neurodevelopmental outcomes in these patients.
Collapse
Affiliation(s)
- Jun Kido
- Department of Pediatrics, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Shirou Matsumoto
- Department of Pediatrics, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Johannes Häberle
- University Children's Hospital Zurich and Children's Research Centre, Zurich, Switzerland
| | - Yoko Nakajima
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Japan
| | - Yoichi Wada
- Department of Pediatrics, Tohoku University School of Medicine, Sendai, Japan
| | - Narutaka Mochizuki
- Department of Neonatal Medicine, Osaka Women's and Children's Hospital, Izumi, Japan
| | - Kei Murayama
- Department of Metabolism, Center for Medical Genetics, Chiba Children's Hospital, Chiba, Japan
| | - Tomoko Lee
- Department of Pediatrics, Hyogo College of Medicine, Nishinomiya, Japan
| | - Hiroshi Mochizuki
- Division of Endocrinology and Metabolism, Saitama Children's Medical Center, Saitama, Japan
| | - Yoriko Watanabe
- Research Institute of Medical Mass Spectrometry, Kurume University School of Medicine, Kurume, Japan
- Department of Pediatrics and Child Health, Kurume University School of Medicine, Kurume, Japan
| | - Reiko Horikawa
- Division of Endocrinology and Metabolism, National Center for Child Health and Development, Tokyo, Japan
| | - Mureo Kasahara
- Organ Transplantation Center, National Center for Child Health and Development, Tokyo, Japan
| | - Kimitoshi Nakamura
- Department of Pediatrics, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
29
|
Kido J, Matsumoto S, Ito T, Hirose S, Fukui K, Kojima-Ishii K, Mushimoto Y, Yoshida S, Ishige M, Sakai N, Nakamura K. Physical, cognitive, and social status of patients with urea cycle disorders in Japan. Mol Genet Metab Rep 2021; 27:100724. [PMID: 33614409 PMCID: PMC7876628 DOI: 10.1016/j.ymgmr.2021.100724] [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] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 01/24/2021] [Indexed: 02/07/2023] Open
Abstract
Urea cycle disorders (UCDs) are inherited metabolic diseases that lead to hyperammonemia. Severe hyperammonemia adversely affects the brain. Therefore, we conducted a nationwide study between January 2000 and March 2018 to understand the present status of UCD patients in Japan regarding diagnosis, treatments, and outcomes. A total of 229 patients with UCDs (126 patients: ornithine transcarbamylase deficiency [OTCD]; 33: carbamoyl phosphate synthetase 1 deficiency [CPS1D]; 48: argininosuccinate synthetase deficiency [ASSD]; 14: argininosuccinate lyase deficiency [ASLD]; and 8: arginase 1 deficiency [ARG1D]) were enrolled in the present study. Although growth impairment is common in patients with UCDs, we discovered that Japanese patients with UCDs were only slightly shorter than the mean height of the general adult population in Japan. Patients with neonatal-onset UCDs are more likely to experience difficulty finding employment and a spouse; however, some patients with late-onset UCDs were employed and married. Additionally, intellectual and developmental disabilities, such as attention deficit hyperactivity disorder (ADHD) and autism, hinder patients with UCDs from achieving a healthy social life. Moreover, we identified that it is vital for patients with UCDs presenting with mild to moderate intellectual disabilities to receive social support. Therefore, we believe the more robust social support system for patients with UCDs may enable them to actively participate in society.
Collapse
Affiliation(s)
- Jun Kido
- Department of Pediatrics, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Shirou Matsumoto
- Department of Pediatrics, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Tetsuya Ito
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Japan
| | - Shinichi Hirose
- Department of Pediatrics, School of Medicine, Fukuoka University, Fukuoka, Japan
| | - Kaori Fukui
- The Department of Pediatrics and Child Health, Kurume University School of Medicine, Kurume, Japan
| | - Kanako Kojima-Ishii
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuichi Mushimoto
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinobu Yoshida
- Department of Pediatrics, Omihachiman Community Medical Center, Shiga, Japan
| | - Mika Ishige
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo, Japan
| | - Norio Sakai
- Child Healthcare and Genetic Science Laboratory, Division of Health Sciences, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kimitoshi Nakamura
- Department of Pediatrics, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
30
|
Wu X, Sun X, Sharma S, Lu Q, Yegambaram M, Hou Y, Wang T, Fineman JR, Black SM. Arginine recycling in endothelial cells is regulated BY HSP90 and the ubiquitin proteasome system. Nitric Oxide 2020; 108:12-19. [PMID: 33338599 DOI: 10.1016/j.niox.2020.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/01/2020] [Accepted: 12/11/2020] [Indexed: 12/26/2022]
Abstract
Despite the saturating concentrations of intracellular l-arginine, nitric oxide (NO) production in endothelial cells (EC) can be stimulated by exogenous arginine. This phenomenon, termed the "arginine paradox" led to the discovery of an arginine recycling pathway in which l-citrulline is recycled to l-arginine by utilizing two important urea cycle enzymes argininosuccinate synthetase (ASS) and argininosuccinate lyase (ASL). Prior work has shown that ASL is present in a NO synthetic complex containing hsp90 and endothelial NO synthase (eNOS). However, it is unclear whether hsp90 forms functional complexes with ASS and ASL and if it is involved regulating their activity. Thus, elucidating the role of hsp90 in the arginine recycling pathway was the goal of this study. Our data indicate that both ASS and ASL are chaperoned by hsp90. Inhibiting hsp90 activity with geldanamycin (GA), decreased the activity of both ASS and ASL and decreased cellular l-arginine levels in bovine aortic endothelial cells (BAEC). hsp90 inhibition led to a time-dependent decrease in ASS and ASL protein, despite no changes in mRNA levels. We further linked this protein loss to a proteasome dependent degradation of ASS and ASL via the E3 ubiquitin ligase, C-terminus of Hsc70-interacting protein (CHIP) and the heat shock protein, hsp70. Transient over-expression of CHIP was sufficient to stimulate ASS and ASL degradation while the over-expression of CHIP mutant proteins identified both TPR- and U-box-domain as essential for ASS and ASL degradation. This study provides a novel insight into the molecular regulation l-arginine recycling in EC and implicates the proteasome pathway as a possible therapeutic target to stimulate NO signaling.
Collapse
Affiliation(s)
- Xiaomin Wu
- Department of Medicine, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, 85721, USA
| | - Xutong Sun
- Department of Medicine, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, 85721, USA
| | - Shruti Sharma
- Center for Biotechnology & Genomic Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Qing Lu
- Department of Medicine, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, 85721, USA
| | - Manivannan Yegambaram
- Department of Medicine, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, 85721, USA
| | - Yali Hou
- Center for Biotechnology & Genomic Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Ting Wang
- Department of Internal Medicine, University of Arizona, Phoenix, AZ, 85004, USA
| | - Jeffrey R Fineman
- The Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA; The Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Stephen M Black
- Department of Medicine, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
31
|
Zhou Y, Eid T, Hassel B, Danbolt NC. Novel aspects of glutamine synthetase in ammonia homeostasis. Neurochem Int 2020; 140:104809. [DOI: 10.1016/j.neuint.2020.104809] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023]
|
32
|
Lerner S, Anderzhanova E, Verbitsky S, Eilam R, Kuperman Y, Tsoory M, Kuznetsov Y, Brandis A, Mehlman T, Mazkereth R, McCarter R, Segal M, Nagamani SCS, Chen A, Erez A. ASL Metabolically Regulates Tyrosine Hydroxylase in the Nucleus Locus Coeruleus. Cell Rep 2020; 29:2144-2153.e7. [PMID: 31747589 PMCID: PMC6902269 DOI: 10.1016/j.celrep.2019.10.043] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/18/2019] [Accepted: 10/10/2019] [Indexed: 12/14/2022] Open
Abstract
Patients with germline mutations in the urea-cycle enzyme argininosuccinate lyase (ASL) are at risk for developing neurobehavioral and cognitive deficits. We find that ASL is prominently expressed in the nucleus locus coeruleus (LC), the central source of norepinephrine. Using natural history data, we show that individuals with ASL deficiency are at risk for developing attention deficits. By generating LC-ASL-conditional knockout (cKO) mice, we further demonstrate altered response to stressful stimuli with increased seizure reactivity in LC-ASL-cKO mice. Depletion of ASL in LC neurons leads to reduced amount and activity of tyrosine hydroxylase (TH) and to decreased catecholamines synthesis, due to decreased nitric oxide (NO) signaling. NO donors normalize catecholamine levels in the LC, seizure sensitivity, and the stress response in LC-ASL-cKO mice. Our data emphasize ASL importance for the metabolic regulation of LC function with translational relevance for ASL deficiency (ASLD) patients as well as for LC-related pathologies. ASL is expressed in the locus coeruleus (LC) and regulates catecholamine synthesis LC-ASL deficiency in mice promotes abnormal stress response and seizure sensitivity LC-ASL deficiency decreases nitric-oxide levels and tyrosine hydroxylase activity NO donors normalize catecholamine production and rescue LC-ASL deficiency phenotype
Collapse
Affiliation(s)
- Shaul Lerner
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Elmira Anderzhanova
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany; Clinic for Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | - Sima Verbitsky
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel; Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Raya Eilam
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Yael Kuperman
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Michael Tsoory
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Yuri Kuznetsov
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Alexander Brandis
- Life Science Core Facility, Weizmann Institute of Science, Rehovot, Israel
| | - Tevie Mehlman
- Life Science Core Facility, Weizmann Institute of Science, Rehovot, Israel
| | - Ram Mazkereth
- The Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | - Robert McCarter
- Center for Translational Sciences, Children's National Health System, The George Washington University, Washington, DC, USA
| | - Menahem Segal
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Sandesh C S Nagamani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Alon Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany; Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
33
|
Wu M, Wang P, Gao M, Shen D, Zhao Q. Transcriptome analysis of the eggs of the silkworm pale red egg (rep-1) mutant at 36 hours after oviposition. PLoS One 2020; 15:e0237242. [PMID: 32764803 PMCID: PMC7413551 DOI: 10.1371/journal.pone.0237242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 07/22/2020] [Indexed: 12/04/2022] Open
Abstract
The egg stage is one of the most critical periods in the life history of silkworms, during which physiological processes such as sex determination, tissue organ formation and differentiation, diapause and pigmentation occur. In addition, egg color gradually emerges around 36h after oviposition. The red egg mutant rep-1, which was recently discovered in the C1(H) wild-type, C1(H) exhibits a brown egg color. In this study, the transcriptome of the eggs was analyzed 36h after oviposition. Between the rep-1 mutant and the C1(H) wild-type, 800 differentially expressed genes (DEGs) were identified, including 325 up-regulated genes and 475 down-regulated genes. These DEGs were mainly involved in biological processes (metabolic process, cellular process, biological regulation and regulation of biological process and localization), cellular components (membrane, membrane part, cell, cell part and organelle) and molecular functions (binding, catalytic activity, transporter activity, structural molecule activity and molecular transducer activity). The pathway enrichment of these DEGs was performed based on the KEGG database, and the results indicated that these DEGs were mainly involved in pathways in the following categories: metabolic pathways, longevity-regulating pathway-multiple species, protein processing in endoplasmic reticulum, peroxisome, carbon metabolism and purine metabolism. Further analysis showed that a large number of silkworm growth- and development-related genes and ommochrome synthesis- and metabolism-related genes were differentially expressed, most of which were up-regulated in the mutant. Our research findings provide new experimental evidence for research on ommochrome pigmentation and lay the foundation for further research on the mechanism of the rep-1 mutant.
Collapse
Affiliation(s)
- Meina Wu
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, China
- The Sericulture Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu, China
| | - Pingyang Wang
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, China
- The Sericulture Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu, China
- Guangxi Zhuang Autonomous Region Research Academy of Sericultural Science, Guangxi, Nanning, China
| | - Mengjie Gao
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, China
- The Sericulture Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu, China
| | - Dongxu Shen
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, China
- The Sericulture Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu, China
| | - Qiaoling Zhao
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, China
- The Sericulture Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu, China
- * E-mail:
| |
Collapse
|
34
|
Del Río C, Martín-Hernández E, Ruiz A, Quijada-Fraile P, Rubio P. Perioperative management of children with urea cycle disorders. Paediatr Anaesth 2020; 30:780-791. [PMID: 32375202 DOI: 10.1111/pan.13905] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Urea cycle disorders are congenital metabolism errors that affect ammonia elimination. Clinical signs and prognosis are strongly influenced by peak ammonia levels. Numerous triggers associated with metabolic decompensation have been described with many of them, including fasting or stress, being related to the perioperative period. AIMS We aimed to assess perioperative complications in pediatric patients with urea cycle disorders requiring general anesthesia in our center. METHODS We reviewed the clinical history of all the pediatric patients with a confirmed urea cycle disorders diagnosis requiring surgery or a diagnostic procedure with anesthesia between January 2002 and June 2018. RESULTS We included 33 operations (major surgery, minor surgery, and diagnostic procedures) carried out on 10 patients via different anesthetic techniques. We observed the following complications: intraoperative hyperglycemia in one case, postoperative vomiting in eight cases, and slightly increased postoperative ammonia levels (54, 59, and 69 µmol/L) with normal preoperative levels in three cases without associated metabolic decompensation. There were two cases of perioperative hyperammonemia (72 and 69 µmol/L) secondary to preoperative metabolic decompensation (137 and 92 µmol/L) with the levels progressively dropping and normalizing in the first 24-48 hours, respectively. CONCLUSIONS Procedures under anesthesia on pediatric patients with urea cycle diseases should be performed by experienced multidisciplinary teams at specialized centers. Perioperative management focused on avoiding catabolism (especially during fasting) and monitoring signs associated with metabolic decompensation to allow for its early treatment should be included in routine anesthetic techniques for children with urea cycle disorders.
Collapse
Affiliation(s)
- Cristina Del Río
- Department of Pediatric Anesthesiology, University Hospital 12 de Octubre, Madrid, Spain
| | - Elena Martín-Hernández
- Unit of Mitochondrial and Inherited Metabolic Diseases, Pediatric Department, University Hospital 12 de Octubre, Madrid, Spain.,National Reference Center, European Reference Network for Hereditary Metabolic Disorders (MetabERN), Madrid, Spain
| | - Alicia Ruiz
- Department of Pediatric Anesthesiology, University Hospital 12 de Octubre, Madrid, Spain
| | - Pilar Quijada-Fraile
- Unit of Mitochondrial and Inherited Metabolic Diseases, Pediatric Department, University Hospital 12 de Octubre, Madrid, Spain.,National Reference Center, European Reference Network for Hereditary Metabolic Disorders (MetabERN), Madrid, Spain
| | - Paloma Rubio
- Department of Pediatric Anesthesiology, University Hospital 12 de Octubre, Madrid, Spain
| |
Collapse
|
35
|
Osawa Y, Wada A, Ohtsu Y, Yamada K, Takizawa T. Late-onset argininosuccinic aciduria associated with hyperammonemia triggered by influenza infection in an adolescent: A case report. Mol Genet Metab Rep 2020; 24:100605. [PMID: 32435591 PMCID: PMC7232106 DOI: 10.1016/j.ymgmr.2020.100605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/05/2020] [Indexed: 12/24/2022] Open
Abstract
Hyperammonemia is a typical symptom of urea cycle disorders. While early-onset argininosuccinic aciduria (ASA) can often be detected by hyperammonemia, patients with late-onset ASA predominantly present with psychomotor retardation and mental disorders. However, in late-onset ASA that develops during early childhood, hyperammonemia can sometimes be caused by acute infections, stress, and reduced dietary intake. Here, we report the case of a 14-year-old boy with late-onset ASA associated with hyperammonemia that was triggered by an influenza A infection. Due to the infection, he presented with a fever and was unable to eat food or take oral medication. He then experienced restlessness, a disturbance in his level of consciousness, and seizures. Hyperammonemia (3286 μg/dL, reference value ≤100 μg/dL) was detected. He was biochemically diagnosed with ASA based on increased serum and urinary argininosuccinic acid levels. Additionally, genetic testing revealed compound heterozygous mutations in the ASL gene: c.91G > A(p.Asp31Asn) and c.1251-1G > C. This case revealed that in late-onset ASA, hyperammonemia can occur not only in early childhood but also during adolescence. Late-onset ASA may have a very broad clinical spectrum that includes hyperammonemia. We suggest that urea cycle disorders such as ASA must be considered when patients present with hyperammonemic decompensation during adolescence.
Collapse
Affiliation(s)
- Yoshimitsu Osawa
- Department of Pediatrics, Gunma University Graduate School of Medicine, Japan.,Department of Pediatrics, Shimane University Faculty of Medicine, Japan
| | - Aya Wada
- Department of Pediatrics, Gunma University Graduate School of Medicine, Japan
| | - Yoshiaki Ohtsu
- Department of Pediatrics, Gunma University Graduate School of Medicine, Japan
| | - Kenji Yamada
- Department of Pediatrics, Shimane University Faculty of Medicine, Japan
| | - Takumi Takizawa
- Department of Pediatrics, Gunma University Graduate School of Medicine, Japan
| |
Collapse
|
36
|
Wang H, Wang X, Qi D, Sun M, Hou Q, Li Y, Jiang H. Establishment of the circadian metabolic phenotype strategy in spontaneously hypertensive rats: a dynamic metabolomics study. J Transl Med 2020; 18:38. [PMID: 31992312 PMCID: PMC6988197 DOI: 10.1186/s12967-020-02222-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 01/10/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Circadian rhythms play a fundamental role in the progression of cardiovascular events. Almost all cardiovascular diseases have a circadian misalignment usually characterized by changes in metabolites. This study aimed to dynamically monitor rhythmic biomarkers, to elucidate the metabolic pathways that are potentially under circadian control in spontaneously hypertensive rats (SHRs), and to eventually establish a circadian metabolic phenotype strategy based on metabolomics. METHODS In this study, an untargeted metabolomics technology was used to dynamically monitor changes in serum metabolites between SHR model group and WKY control group. Liquid chromatography-mass spectrometry (LC-MS) combined with multivariate statistical analysis was applied to identify markers of hypertension rhythm imbalance. The concentrations of amino acids and their metabolites identified as markers were quantified by a subsequent targeted metabolomics analysis. Overall, these approaches comprehensively explored the rhythm mechanism and established a circadian metabolic phenotype strategy. RESULTS The metabolic profile revealed a disorder in the diurnal metabolism pattern in SHRs. Moreover, multivariate statistical analysis revealed metabolic markers of rhythm homeostasis, such as arginine, proline, phenylalanine, citric acid, L-malic acid, succinic acid, etc., accompanied by an imbalance in hypertension. The key metabolic pathways related to rhythm imbalance in hypertension were found by enrichment analysis, including amino acid metabolism, and the tricarboxylic acid cycle (TCA). In addition, the quantitative analysis of amino acids and their metabolites showed that the changes in leucine, isoleucine, valine, taurine, serine, and glycine were the most obvious. CONCLUSIONS In summary, this study illustrated the relationship between metabolites and the pathways across time on hypertension. These results may provide a theoretical basis for personalized treatment programmes and timing for hypertension.
Collapse
Affiliation(s)
- Huanjun Wang
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Xiaoming Wang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China.,Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China.,Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Dongmei Qi
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China.,Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China.,Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Mengjia Sun
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Qingqing Hou
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Yunlun Li
- TCM Clinical Research Base for Hypertension, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China.
| | - Haiqiang Jiang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China. .,Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China. .,Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China.
| |
Collapse
|
37
|
Stettner N, Rosen C, Bernshtein B, Gur-Cohen S, Frug J, Silberman A, Sarver A, Carmel-Neiderman NN, Eilam R, Biton I, Pevsner-Fischer M, Zmora N, Brandis A, Bahar Halpern K, Mazkereth R, di Bernardo D, Brunetti-Pierri N, Premkumar MH, Dank G, Nagamani SCS, Jung S, Harmelin A, Erez A. Induction of Nitric-Oxide Metabolism in Enterocytes Alleviates Colitis and Inflammation-Associated Colon Cancer. Cell Rep 2019; 23:1962-1976. [PMID: 29768197 PMCID: PMC5976577 DOI: 10.1016/j.celrep.2018.04.053] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 02/05/2018] [Accepted: 04/12/2018] [Indexed: 12/30/2022] Open
Abstract
Nitric oxide (NO) plays an established role in numerous physiological and pathological processes, but the specific cellular sources of NO in disease pathogenesis remain unclear, preventing the implementation of NO-related therapy. Argininosuccinate lyase (ASL) is the only enzyme able to produce arginine, the substrate for NO generation by nitric oxide synthase (NOS) isoforms. Here, we generated cell-specific conditional ASL knockout mice in combination with genetic and chemical colitis models. We demonstrate that NO derived from enterocytes alleviates colitis by decreasing macrophage infiltration and tissue damage, whereas immune cell-derived NO is associated with macrophage activation, resulting in increased severity of inflammation. We find that induction of endogenous NO production by enterocytes with supplements that upregulate ASL expression and complement its substrates results in improved epithelial integrity and alleviation of colitis and of inflammation-associated colon cancer.
Collapse
Affiliation(s)
- Noa Stettner
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel; Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel; Koret School of Veterinary Medicine, Hebrew University, Rehovot, Israel
| | - Chava Rosen
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel; The Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel
| | - Biana Bernshtein
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Shiri Gur-Cohen
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Julia Frug
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Silberman
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Alona Sarver
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | | | - Raya Eilam
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Inbal Biton
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | | | - Niv Zmora
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Alexander Brandis
- Department of Biological Services, Weizmann Institute of Science, Rehovot, Israel
| | - Keren Bahar Halpern
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ram Mazkereth
- The Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Diego di Bernardo
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy; Department of Chemical, Materials and Industrial Engineering, Federico II University, Naples, Italy
| | - Nicola Brunetti-Pierri
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy; Department of Translational Medicine, Federico II University, Naples, Italy
| | - Muralidhar H Premkumar
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Gillian Dank
- Koret School of Veterinary Medicine, Hebrew University, Rehovot, Israel
| | - Sandesh C S Nagamani
- Texas Children's Hospital, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Harmelin
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
38
|
Free Radical Scavengers Prevent Argininosuccinic Acid-Induced Oxidative Stress in the Brain of Developing Rats: a New Adjuvant Therapy for Argininosuccinate Lyase Deficiency? Mol Neurobiol 2019; 57:1233-1244. [PMID: 31707633 DOI: 10.1007/s12035-019-01825-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 10/24/2019] [Indexed: 12/31/2022]
Abstract
Tissue accumulation and high urinary excretion of argininosuccinate (ASA) is the biochemical hallmark of argininosuccinate lyase deficiency (ASLD), a urea cycle disorder mainly characterized by neurologic abnormalities, whose pathogenesis is still unknown. Thus, in the present work, we evaluated the in vitro and in vivo effects of ASA on a large spectrum of oxidative stress parameters in brain of adolescent rats in order to test whether disruption of redox homeostasis could be involved in neurodegeneration of this disorder. ASA provoked in vitro lipid and protein oxidation, decreased reduced glutathione (GSH) concentrations, and increased reactive oxygen species generation in cerebral cortex and striatum. Furthermore, these effects were totally prevented or attenuated by the antioxidants melatonin and GSH. Similar results were obtained by intrastriatal administration of ASA, in addition to increased reactive nitrogen species generation and decreased activities of superoxide dismutase, glutathione peroxidase, and glutathione S-transferase. It was also observed that melatonin and N-acetylcysteine prevented most of ASA-induced in vivo pro-oxidant effects in striatum. Taken together, these data indicate that disturbance of redox homeostasis induced at least in part by high brain ASA concentrations per se may potentially represent an important pathomechanism of neurodegeneration in patients with ASLD and that therapeutic trials with appropriate antioxidants may be an adjuvant treatment for these patients.
Collapse
|
39
|
Diez-Fernandez C, Hertig D, Loup M, Diserens G, Henry H, Vermathen P, Nuoffer JM, Häberle J, Braissant O. Argininosuccinate neurotoxicity and prevention by creatine in argininosuccinate lyase deficiency: An in vitro study in rat three-dimensional organotypic brain cell cultures. J Inherit Metab Dis 2019; 42:1077-1087. [PMID: 30907007 DOI: 10.1002/jimd.12090] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 03/04/2019] [Accepted: 03/22/2019] [Indexed: 12/31/2022]
Abstract
The urea cycle disorder (UCD) argininosuccinate lyase (ASL) deficiency, caused by a defective ASL enzyme, exhibits a wide range of phenotypes, from life-threatening neonatal hyperammonemia to asymptomatic patients, with only the biochemical marker argininosuccinic acid (ASA) elevated in body fluids. Remarkably, even without ever suffering from hyperammonemia, patients often develop severe cognitive impairment and seizures. The goal of this study was to understand the effect on the known toxic metabolite ASA and the assumed toxic metabolite guanidinosuccinic acid (GSA) on developing brain cells, and to evaluate the potential role of creatine (Cr) supplementation, as it was described protective for brain cells exposed to ammonia. We used an in vitro model, in which we exposed three-dimensional (3D) organotypic rat brain cell cultures in aggregates to different combinations of the metabolites of interest at two time points (representing two different developmental stages). After harvest and cryopreservation of the cell cultures, the samples were analyzed mainly by metabolite analysis, immunohistochemistry, and western blotting. ASA and GSA were found toxic for astrocytes and neurons. This toxicity could be reverted in vitro by Cr. As well, an antiapoptotic effect of ASA was revealed, which could contribute to the neurotoxicity in ASL deficiency. Further studies in human ASL deficiency will be required to understand the biochemical situation in the brain of affected patients, and to investigate the impact of high or low arginine doses on brain Cr availability. In addition, clinical trials to evaluate the beneficial effect of Cr supplementation in ASL deficiency would be valuable.
Collapse
Affiliation(s)
- Carmen Diez-Fernandez
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Damian Hertig
- Division of Pediatric Endocrinology, Diabetology and Metabolism and University Institute of Clinical Chemistry, Inselspital, University Hospital, University of Bern, Bern, Switzerland
- AMSM, Department of Biomedical Research, University of Bern, Bern, Switzerland
- AMSM, Department of Radiology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Marc Loup
- Service of Clinical Chemistry, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Gaelle Diserens
- AMSM, Department of Biomedical Research, University of Bern, Bern, Switzerland
- AMSM, Department of Radiology, University of Bern, Bern, Switzerland
| | - Hugues Henry
- Service of Clinical Chemistry, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Peter Vermathen
- AMSM, Department of Biomedical Research, University of Bern, Bern, Switzerland
- AMSM, Department of Radiology, University of Bern, Bern, Switzerland
| | - Jean-Marc Nuoffer
- Division of Pediatric Endocrinology, Diabetology and Metabolism and University Institute of Clinical Chemistry, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| | - Johannes Häberle
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Olivier Braissant
- Service of Clinical Chemistry, Lausanne University Hospital and University of Lausanne, Switzerland
| |
Collapse
|
40
|
Baruteau J, Diez-Fernandez C, Lerner S, Ranucci G, Gissen P, Dionisi-Vici C, Nagamani S, Erez A, Häberle J. Argininosuccinic aciduria: Recent pathophysiological insights and therapeutic prospects. J Inherit Metab Dis 2019; 42:1147-1161. [PMID: 30723942 DOI: 10.1002/jimd.12047] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 12/20/2018] [Indexed: 12/30/2022]
Abstract
The first patients affected by argininosuccinic aciduria (ASA) were reported 60 years ago. The clinical presentation was initially described as similar to other urea cycle defects, but increasing evidence has shown overtime an atypical systemic phenotype with a paradoxical observation, that is, a higher rate of neurological complications contrasting with a lower rate of hyperammonaemic episodes. The disappointing long-term clinical outcomes of many of the patients have challenged the current standard of care and therapeutic strategy, which aims to normalize plasma ammonia and arginine levels. Interrogations have raised about the benefit of newborn screening or liver transplantation on the neurological phenotype. Over the last decade, novel discoveries enabled by the generation of new transgenic argininosuccinate lyase (ASL)-deficient mouse models have been achieved, such as, a better understanding of ASL and its close interaction with nitric oxide metabolism, ASL physiological role outside the liver, and the pathophysiological role of oxidative/nitrosative stress or excessive arginine treatment. Here, we present a collaborative review, which highlights these recent discoveries and novel emerging concepts about ASL role in human physiology, ASA clinical phenotype and geographic prevalence, limits of current standard of care and newborn screening, pathophysiology of the disease, and emerging novel therapies. We propose recommendations for monitoring of ASA patients. Ongoing research aims to better understand the underlying pathogenic mechanisms of the systemic disease to design novel therapies.
Collapse
Affiliation(s)
- Julien Baruteau
- UCL Great Ormond Street Institute of Child Health, NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
- Metabolic Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Carmen Diez-Fernandez
- Division of Metabolism and Children Research Centre (CRC), University Children's Hospital, Zurich, Switzerland
| | - Shaul Lerner
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israël
| | - Giusy Ranucci
- Division of Metabolism, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Paul Gissen
- UCL Great Ormond Street Institute of Child Health, NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
- Metabolic Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Carlo Dionisi-Vici
- Division of Metabolism, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Sandesh Nagamani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israël
| | - Johannes Häberle
- Division of Metabolism and Children Research Centre (CRC), University Children's Hospital, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP) and Neuroscience Center Zurich (ZNZ), Zurich, Switzerland
| |
Collapse
|
41
|
Häberle J, Burlina A, Chakrapani A, Dixon M, Karall D, Lindner M, Mandel H, Martinelli D, Pintos-Morell G, Santer R, Skouma A, Servais A, Tal G, Rubio V, Huemer M, Dionisi-Vici C. Suggested guidelines for the diagnosis and management of urea cycle disorders: First revision. J Inherit Metab Dis 2019; 42:1192-1230. [PMID: 30982989 DOI: 10.1002/jimd.12100] [Citation(s) in RCA: 277] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 02/06/2023]
Abstract
In 2012, we published guidelines summarizing and evaluating late 2011 evidence for diagnosis and therapy of urea cycle disorders (UCDs). With 1:35 000 estimated incidence, UCDs cause hyperammonemia of neonatal (~50%) or late onset that can lead to intellectual disability or death, even while effective therapies do exist. In the 7 years that have elapsed since the first guideline was published, abundant novel information has accumulated, experience on newborn screening for some UCDs has widened, a novel hyperammonemia-causing genetic disorder has been reported, glycerol phenylbutyrate has been introduced as a treatment, and novel promising therapeutic avenues (including gene therapy) have been opened. Several factors including the impact of the first edition of these guidelines (frequently read and quoted) may have increased awareness among health professionals and patient families. However, under-recognition and delayed diagnosis of UCDs still appear widespread. It was therefore necessary to revise the original guidelines to ensure an up-to-date frame of reference for professionals and patients as well as for awareness campaigns. This was accomplished by keeping the original spirit of providing a trans-European consensus based on robust evidence (scored with GRADE methodology), involving professionals on UCDs from nine countries in preparing this consensus. We believe this revised guideline, which has been reviewed by several societies that are involved in the management of UCDs, will have a positive impact on the outcomes of patients by establishing common standards, and spreading and harmonizing good practices. It may also promote the identification of knowledge voids to be filled by future research.
Collapse
Affiliation(s)
- Johannes Häberle
- University Children's Hospital Zurich and Children's Research Centre, Zurich, Switzerland
| | - Alberto Burlina
- Division of Inborn Metabolic Disease, Department of Pediatrics, University Hospital Padua, Padova, Italy
| | - Anupam Chakrapani
- Department of Metabolic Medicine, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Marjorie Dixon
- Dietetics, Great Ormond Street Hospital for Children, NHS Trust, London, UK
| | - Daniela Karall
- Clinic for Pediatrics, Division of Inherited Metabolic Disorders, Medical University of Innsbruck, Innsbruck, Austria
| | - Martin Lindner
- University Children's Hospital, Frankfurt am Main, Germany
| | - Hanna Mandel
- Institute of Human Genetics and metabolic disorders, Western Galilee Medical Center, Nahariya, Israel
| | - Diego Martinelli
- Division of Metabolism, Bambino Gesù Children's Hospital, Rome, Italy
| | - Guillem Pintos-Morell
- Centre for Rare Diseases, University Hospital Vall d'Hebron, Barcelona, Spain
- CIBERER_GCV08, Research Institute IGTP, Barcelona, Spain
- Universitat Autònoma de Barcelona, Barcelona, Spain
| | - René Santer
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anastasia Skouma
- Institute of Child Health, Agia Sofia Children's Hospital, Athens, Greece
| | - Aude Servais
- Service de Néphrologie et maladies métaboliques adulte Hôpital Necker 149, Paris, France
| | - Galit Tal
- The Ruth Rappaport Children's Hospital, Rambam Medical Center, Haifa, Israel
| | - Vicente Rubio
- Instituto de Biomedicina de Valencia (IBV-CSIC), Centro de Investigación Biomédica en Red para Enfermedades Raras (CIBERER), Valencia, Spain
| | - Martina Huemer
- University Children's Hospital Zurich and Children's Research Centre, Zurich, Switzerland
- Department of Paediatrics, Landeskrankenhaus Bregenz, Bregenz, Austria
| | | |
Collapse
|
42
|
Dysregulation in the Brain Protein Profile of Zebrafish Lacking the Parkinson’s Disease-Related Protein DJ-1. Mol Neurobiol 2019; 56:8306-8322. [DOI: 10.1007/s12035-019-01667-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/31/2019] [Indexed: 02/07/2023]
|
43
|
Whole-Exome Sequencing Identified a Novel Compound Heterozygous Genotype in ASL in a Chinese Han Patient with Argininosuccinate Lyase Deficiency. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3530198. [PMID: 31183366 PMCID: PMC6515145 DOI: 10.1155/2019/3530198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 01/03/2019] [Accepted: 03/07/2019] [Indexed: 11/17/2022]
Abstract
Pathogenic variants in the argininosuccinate lyase (ASL) gene have been shown to cause argininosuccinate lyase deficiency (ASLD); therefore, sequencing analysis offers advantages for prenatal testing and counseling in families afflicted with this condition. Here, we performed a genetic analysis of an ASLD patient and his family with an aim to offer available information for clinical diagnosis. The research subjects were a 23-month-old patient with a high plasma level of citrulline and his unaffected parents. Whole-exome sequencing identified potential related ASL gene mutations in this trio. Enzymatic activity was detected spectrophotometrically by a coupled assay using arginase and measuring urea production. We identified a novel nonsynonymous mutation (c.206A>G, p.Lys69Arg) and a stop mutation (c.637C>T, p.Arg213∗) in ASL in a Chinese Han patient with ASLD. The enzymatic activity of a p.Lys69Arg ASL construct in human embryonic kidney 293T cells was significantly reduced compared to that of the wild-type construct, and no significant activity was observed for the p.Arg213∗ construct. Compound heterozygous p.Lys69Arg and p.Arg213∗ mutations that resulted in reduced ASL enzyme activity were found in a patient with ASLD. This finding expands the clinical spectrum of ASL pathogenic variants.
Collapse
|
44
|
Wang Y, Sun Y, Liu M, Zhang X, Jiang T. Functional Characterization of Argininosuccinate Lyase Gene Variants by Mini-Gene Splicing Assay. Front Genet 2019; 10:436. [PMID: 31156699 PMCID: PMC6533879 DOI: 10.3389/fgene.2019.00436] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 04/29/2019] [Indexed: 11/13/2022] Open
Abstract
Objective Argininosuccinate lyase (ASL) gene mutations account for argininosuccinic aciduria (ASA). This study aimed to design a minigene construct of ASL gene in order to investigate the impact of variants on splicing. Methods The peripheral blood samples were collected from the family members, and genomic DNA was extracted for gene diagnosis using the total exon sequencing method. The novel mutation gene was cloned into pEGFP-C1 vector, and the pathogenicity of the mutation was examined in cultured cells in vitro. Results The clinical diagnosis of the proband as ASA was clear. Two pathogenic mutations, c.281G>T (p.Arg94Leu) and c.208-15 T>A were detected in the ASL gene, and the two mutations had not been reported. The minigene expression in vitro confirmed that c.208-15 T>A could cause aberrant splicing, resulting in the retention of 13 bp in intron 2 to exon 3. Conclusion Two new pathogenic mutations of ASL gene, c.208-15 T>A and c.281G>T, were found in an ASA family, which enriches the mutational profile of the ASL gene and provides a basis for genetic diagnosis of ASA. Minigenes are optimal approaches to determine whether the intron mutation can cause aberrant splicing.
Collapse
Affiliation(s)
- Yanyun Wang
- Center of Genetic Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Yun Sun
- Center of Genetic Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Ming Liu
- Center of Child Health Care, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Xiaojuan Zhang
- Center of Genetic Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Tao Jiang
- Center of Genetic Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China.,Center of Genetic Medicine, The Affiliated Obstetrics and Gynecology Hospital with Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| |
Collapse
|
45
|
Pankowicz FP, Barzi M, Kim KH, Legras X, Martins CS, Wooton-Kee CR, Lagor WR, Marini JC, Elsea SH, Bissig-Choisat B, Moore DD, Bissig KD. Rapid Disruption of Genes Specifically in Livers of Mice Using Multiplex CRISPR/Cas9 Editing. Gastroenterology 2018; 155:1967-1970.e6. [PMID: 30170115 PMCID: PMC6420307 DOI: 10.1053/j.gastro.2018.08.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 08/17/2018] [Accepted: 08/22/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS Despite advances in gene editing technologies, generation of tissue-specific knockout mice is time-consuming. We used CRISPR/Cas9-mediated genome editing to disrupt genes in livers of adult mice in just a few months, which we refer to as somatic liver knockouts. METHODS In this system, Fah-/- mice are given hydrodynamic tail vein injections of plasmids carrying CRISPR/Cas9 designed to excise exons in Hpd; the Hpd-edited hepatocytes have a survival advantage in these mice. Plasmids that target Hpd and a separate gene of interest can therefore be used to rapidly generate mice with liver-specific deletion of nearly any gene product. RESULTS We used this system to create mice with liver-specific knockout of argininosuccinate lyase, which develop hyperammonemia, observed in humans with mutations in this gene. We also created mice with liver-specific knockout of ATP binding cassette subfamily B member 11, which encodes the bile salt export pump. We found that these mice have a biochemical phenotype similar to that of Abcb11-/- mice. We then used this system to knock out expression of 5 different enzymes involved in drug metabolism within the same mouse. CONCLUSIONS This approach might be used to develop new models of liver diseases and study liver functions of genes that are required during development.
Collapse
Affiliation(s)
- Francis P Pankowicz
- Center for Cell and Gene Therapy, Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Mercedes Barzi
- Center for Cell and Gene Therapy, Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Kang Ho Kim
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Xavier Legras
- Center for Cell and Gene Therapy, Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Celeste Santos Martins
- Center for Cell and Gene Therapy, Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Clavia Ruth Wooton-Kee
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - William R. Lagor
- Center for Cell and Gene Therapy, Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas
| | - Juan C Marini
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Sarah H Elsea
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston
| | - Beatrice Bissig-Choisat
- Center for Cell and Gene Therapy, Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - David D Moore
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, USA,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Karl-Dimiter Bissig
- Center for Cell and Gene Therapy, Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, Texas; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
46
|
Savy N, Brossier D, Brunel-Guitton C, Ducharme-Crevier L, Du Pont-Thibodeau G, Jouvet P. Acute pediatric hyperammonemia: current diagnosis and management strategies. Hepat Med 2018; 10:105-115. [PMID: 30254497 PMCID: PMC6140721 DOI: 10.2147/hmer.s140711] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Acute hyperammonemia may induce a neurologic impairment leading to an acute life-threatening condition. Coma duration, ammonia peak level, and hyperammonemia duration are the main risk factors of hyperammonemia-related neurologic deficits and death. In children, hyperammonemia is mainly caused by severe liver failure and inborn errors of metabolism. In an acute setting, obtaining reliable plasma ammonia levels can be challenging because of the preanalytical difficulties that need to be addressed carefully. The management of hyperammonemia includes 1) identification of precipitating factors and cerebral edema presence, 2) a decrease in ammonia production by reducing protein intake and reversing catabolism, and 3) ammonia removal with pharmacologic treatment and, in the most severe cases, with extracorporeal therapies. In case of severe coma, transcranial Doppler ultrasound could be the method of choice to noninvasively monitor cerebral blood flow and titrate therapies.
Collapse
Affiliation(s)
- Nadia Savy
- Department of Pediatrics, CHU Sainte-Justine, University of Montreal, Montreal, QC, Canada,
| | - David Brossier
- Department of Pediatrics, Pediatric Intensive Care Unit, CHU Caen, Caen, France
| | | | | | | | - Philippe Jouvet
- Department of Pediatrics, CHU Sainte-Justine, University of Montreal, Montreal, QC, Canada,
| |
Collapse
|
47
|
Nault JC, Couchy G, Caruso S, Meunier L, Caruana L, Letouzé E, Rebouissou S, Paradis V, Calderaro J, Zucman-Rossi J. Argininosuccinate synthase 1 and periportal gene expression in sonic hedgehog hepatocellular adenomas. Hepatology 2018; 68:964-976. [PMID: 29572896 DOI: 10.1002/hep.29884] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/29/2018] [Accepted: 02/28/2018] [Indexed: 12/25/2022]
Abstract
UNLABELLED Genetic alterations define different molecular subclasses of hepatocellular adenoma (HCA) linked with risk factors, histology and clinical behavior. Recently, Argininosuccinate Synthase 1 (ASS1), a major periportal protein, was proposed as a marker of HCA with a high risk of hemorrhage. We aimed to assess the significance of ASS1 expression through the scope of the HCA molecular classification. ASS1 expression was evaluated using RNAseq, quantitative reverse transcriptase polymerase chain reaction (RT-PCR) and Immunohistochemistry. ASS1 and glioma-associated oncogene 1 (GLI1) expression were analyzed in vitro after modulation of GLI1 expression. Using RNAseq in 27 HCA and five nontumor liver samples, ASS1 expression was highly correlated with GLI1 expression (P<0.0001, R=0.75). In the overall series of 408 HCA, ASS1 overexpression was significantly associated with sonic hedgehog HCA (shHCA) compared to other molecular subgroups (P<0.0001), suggesting that sonic hedgehog signaling controls ASS1 expression. GLI1 expression silencing by siRNA induced a downregulation of ASS1 in PLC/PFR5 and SNU878 cell lines. In 390 HCA, we showed that ASS1 expression belonged to the periportal expression program that was maintained in shHCA but down-regulated in all the other HCA subtypes. In contrast, HCA with β-catenin activation showed an activation of a perivenous program. Despite the significant association between GLI1 and ASS1 expression, ASS1 mRNA expression was not associated with specific clinical features. At the protein level using immunohistochemistry, prostaglandin D synthase (PTGDS) was strongly and specifically overexpressed in shHCA. CONCLUSION ASS1 is associated with sonic hedgehog activation as part of a periportal program expressed in shHCA, a molecular subgroup defined by INHBE-GLI1 gene fusion. (Hepatology 2018).
Collapse
Affiliation(s)
- Jean-Charles Nault
- Inserm UMR-1162, Génomique fonctionnelle des Tumeurs solides, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, Paris, France.,Liver unit, Hôpital Jean Verdier, Hôpitaux Universitaires Paris-Seine-Saint-Denis, Assistance-Publique Hôpitaux de Paris, APHP, Bondy, France.,Unité de Formation et de Recherche Santé Médecine et Biologie Humaine, Université Paris 13, Communauté d'Universités et Etablissements Sorbonne Paris Cité, Paris, France
| | - Gabrielle Couchy
- Inserm UMR-1162, Génomique fonctionnelle des Tumeurs solides, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, Paris, France
| | - Stefano Caruso
- Inserm UMR-1162, Génomique fonctionnelle des Tumeurs solides, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, Paris, France
| | - Léa Meunier
- Inserm UMR-1162, Génomique fonctionnelle des Tumeurs solides, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, Paris, France
| | - Laure Caruana
- Inserm UMR-1162, Génomique fonctionnelle des Tumeurs solides, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, Paris, France
| | - Eric Letouzé
- Inserm UMR-1162, Génomique fonctionnelle des Tumeurs solides, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, Paris, France
| | - Sandra Rebouissou
- Inserm UMR-1162, Génomique fonctionnelle des Tumeurs solides, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, Paris, France
| | - Valérie Paradis
- Service d'anatomopathologie, Hôpital Beaujon, Clichy, Centre de Recherche sur l'inflammation, UMR 1149, INSERM-Paris Diderot University, Paris, France
| | - Julien Calderaro
- Inserm UMR-1162, Génomique fonctionnelle des Tumeurs solides, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, Paris, France.,Service d'anatomopathologie, Hôpital Henri Mondor, Créteil; Université Paris Est Créteil, Inserm U955, Team 18, Institut Mondor de Recherche Biomédicale, Paris, France
| | - Jessica Zucman-Rossi
- Inserm UMR-1162, Génomique fonctionnelle des Tumeurs solides, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, Paris, France.,Hôpital Europeen Georges Pompidou, HEGP, F-75015, Assistance Publique-Hôpitaux de Paris, APHP, Paris, France
| |
Collapse
|
48
|
Baruteau J, Perocheau DP, Hanley J, Lorvellec M, Rocha-Ferreira E, Karda R, Ng J, Suff N, Diaz JA, Rahim AA, Hughes MP, Banushi B, Prunty H, Hristova M, Ridout DA, Virasami A, Heales S, Howe SJ, Buckley SMK, Mills PB, Gissen P, Waddington SN. Argininosuccinic aciduria fosters neuronal nitrosative stress reversed by Asl gene transfer. Nat Commun 2018; 9:3505. [PMID: 30158522 PMCID: PMC6115417 DOI: 10.1038/s41467-018-05972-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/06/2018] [Indexed: 12/26/2022] Open
Abstract
Argininosuccinate lyase (ASL) belongs to the hepatic urea cycle detoxifying ammonia, and the citrulline-nitric oxide (NO) cycle producing NO. ASL-deficient patients present argininosuccinic aciduria characterised by hyperammonaemia, multiorgan disease and neurocognitive impairment despite treatment aiming to normalise ammonaemia without considering NO imbalance. Here we show that cerebral disease in argininosuccinic aciduria involves neuronal oxidative/nitrosative stress independent of hyperammonaemia. Intravenous injection of AAV8 vector into adult or neonatal ASL-deficient mice demonstrates long-term correction of the hepatic urea cycle and the cerebral citrulline-NO cycle, respectively. Cerebral disease persists if ammonaemia only is normalised but is dramatically reduced after correction of both ammonaemia and neuronal ASL activity. This correlates with behavioural improvement and reduced cortical cell death. Thus, neuronal oxidative/nitrosative stress is a distinct pathophysiological mechanism from hyperammonaemia. Disease amelioration by simultaneous brain and liver gene transfer with one vector, to treat both metabolic pathways, provides new hope for hepatocerebral metabolic diseases.
Collapse
Affiliation(s)
- Julien Baruteau
- Gene Transfer Technology Group, Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
- Metabolic Medicine Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, WC1N 3JH, UK
- Genetics and Genomic Medicine Programme, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Dany P Perocheau
- Gene Transfer Technology Group, Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
| | - Joanna Hanley
- Genetics and Genomic Medicine Programme, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Maëlle Lorvellec
- Genetics and Genomic Medicine Programme, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Eridan Rocha-Ferreira
- Perinatal Brain Repair Group, Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
| | - Rajvinder Karda
- Gene Transfer Technology Group, Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
| | - Joanne Ng
- Gene Transfer Technology Group, Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
- Neurology Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, WC1N 3JH, UK
| | - Natalie Suff
- Gene Transfer Technology Group, Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
| | - Juan Antinao Diaz
- Gene Transfer Technology Group, Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
| | - Ahad A Rahim
- Department of Pharmacology, School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Michael P Hughes
- Department of Pharmacology, School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Blerida Banushi
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Helen Prunty
- Department of Paediatric Laboratory Medicine, Great Ormond Street Hospital for Children NHS Foundation Trust, London, WC1N 3JH, UK
| | - Mariya Hristova
- Perinatal Brain Repair Group, Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
| | - Deborah A Ridout
- Population, Policy and Practice Programme, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1E, UK
| | - Alex Virasami
- Histopathology Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, WC1N 3JH, UK
| | - Simon Heales
- Genetics and Genomic Medicine Programme, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
- Department of Paediatric Laboratory Medicine, Great Ormond Street Hospital for Children NHS Foundation Trust, London, WC1N 3JH, UK
| | - Stewen J Howe
- Gene Transfer Technology Group, Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
| | - Suzanne M K Buckley
- Gene Transfer Technology Group, Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
| | - Philippa B Mills
- Genetics and Genomic Medicine Programme, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Paul Gissen
- Metabolic Medicine Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, WC1N 3JH, UK
- Genetics and Genomic Medicine Programme, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Simon N Waddington
- Gene Transfer Technology Group, Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK.
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witswatersrand, Johannesburg, South Africa.
| |
Collapse
|
49
|
Pathway Analysis of a Transcriptome and Metabolite Profile to Elucidate a Compensatory Mechanism for Taurine Deficiency in the Heart of Taurine Transporter Knockout Mice. J 2018. [DOI: 10.3390/j1010007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Taurine, which is abundant in mammalian tissues, especially in the heart, is essential for cellular osmoregulation. We previously reported that taurine deficiency leads to changes in the levels of several metabolites, suggesting that alterations in those metabolites might compensate in part for tissue taurine loss, a process that would be important in maintaining cardiac homeostasis. In this study, we investigated the molecular basis for changes in the metabolite profile of a taurine-deficient heart using pathway analysis based on the transcriptome and metabolome profile in the hearts of taurine transporter knockout mice (TauTKO mice), which have been reported by us. First, the genes associated with transport activity, such as the solute carrier (SLC) family, are increased in TauTKO mice, while the established transporters for metabolites that are elevated in the TauTKO heart, such as betaine and carnitine, are not altered by taurine deficiency. Second, the integrated analysis using transcriptome and metabolome data revealed significant increases and/or decreases in the genes involved in Arginine metabolism, Ketone body degradation, Glycerophospholipid metabolism, and Fatty acid metabolism in the KEGG pathway database. In conclusion, these pathway analyses revealed genetic compensatory mechanisms involved in the control of the metabolome profile of the taurine-deficient heart.
Collapse
|
50
|
Kim D, Ko JM, Kim YM, Seo GH, Kim GH, Lee BH, Yoo HW. Low prevalence of argininosuccinate lyase deficiency among inherited urea cycle disorders in Korea. J Hum Genet 2018; 63:911-917. [DOI: 10.1038/s10038-018-0467-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/04/2018] [Accepted: 04/26/2018] [Indexed: 12/31/2022]
|