1
|
Duff C, Alexander IE, Baruteau J. Gene therapy for urea cycle defects: An update from historical perspectives to future prospects. J Inherit Metab Dis 2024; 47:50-62. [PMID: 37026568 PMCID: PMC10953416 DOI: 10.1002/jimd.12609] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/08/2023]
Abstract
Urea cycle defects (UCDs) are severe inherited metabolic diseases with high unmet needs which present a permanent risk of hyperammonaemic decompensation and subsequent acute death or neurological sequelae, when treated with conventional dietetic and medical therapies. Liver transplantation is currently the only curative option, but has the potential to be supplanted by highly effective gene therapy interventions without the attendant need for life-long immunosuppression or limitations imposed by donor liver supply. Over the last three decades, pioneering genetic technologies have been explored to circumvent the consequences of UCDs, improve quality of life and long-term outcomes: adenoviral vectors, adeno-associated viral vectors, gene editing, genome integration and non-viral technology with messenger RNA. In this review, we present a summarised view of this historical path, which includes some seminal milestones of the gene therapy's epic. We provide an update about the state of the art of gene therapy technologies for UCDs and the current advantages and pitfalls driving future directions for research and development.
Collapse
Affiliation(s)
- Claire Duff
- Genetics and Genomic Medicine Department, Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
| | - Ian E. Alexander
- Gene Therapy Research Unit, Children's Medical Research Institute, Faculty of Medicine and HealthThe University of Sydney and Sydney Children's Hospitals NetworkWestmeadNew South WalesAustralia
- Discipline of Child and Adolescent HealthThe University of SydneyWestmeadNew South WalesAustralia
| | - Julien Baruteau
- Genetics and Genomic Medicine Department, Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
- National Institute of Health Research Great Ormond Street Biomedical Research CentreLondonUK
- Metabolic Medicine DepartmentGreat Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| |
Collapse
|
2
|
Diaz GA, Bechter M, Cederbaum SD. The role and control of arginine levels in arginase 1 deficiency. J Inherit Metab Dis 2023; 46:3-14. [PMID: 36175366 PMCID: PMC10091968 DOI: 10.1002/jimd.12564] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 01/19/2023]
Abstract
Arginase 1 Deficiency (ARG1-D) is a rare urea cycle disorder that results in persistent hyperargininemia and a distinct, progressive neurologic phenotype involving developmental delay, intellectual disability, and spasticity, predominantly affecting the lower limbs and leading to mobility impairment. Unlike the typical presentation of other urea cycle disorders, individuals with ARG1-D usually appear healthy at birth and hyperammonemia is comparatively less severe and less common. Clinical manifestations typically begin to develop in early childhood in association with high plasma arginine levels, with hyperargininemia (and not hyperammonemia) considered to be the primary driver of disease sequelae. Nearly five decades of clinical experience with ARG1-D and empirical studies in genetically manipulated models have generated a large body of evidence that, when considered in aggregate, implicates arginine directly in disease pathophysiology. Severe dietary protein restriction to minimize arginine intake and diversion of ammonia from the urea cycle are the mainstay of care. Although this approach does reduce plasma arginine and improve patients' cognitive and motor/mobility manifestations, it is inadequate to achieve and maintain sufficiently low arginine levels and prevent progression in the long term. This review presents a comprehensive discussion of the clinical and scientific literature, the effects and limitations of the current standard of care, and the authors' perspectives regarding the past, current, and future management of ARG1-D.
Collapse
Affiliation(s)
- George A Diaz
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Stephen D Cederbaum
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
3
|
Bazo A, Lantero A, Mauleón I, Neri L, Poms M, Häberle J, Ricobaraza A, Bénichou B, Combal JP, Gonzalez-Aseguinolaza G, Aldabe R. Gene Therapy in Combination with Nitrogen Scavenger Pretreatment Corrects Biochemical and Behavioral Abnormalities of Infant Citrullinemia Type 1 Mice. Int J Mol Sci 2022; 23:14940. [PMID: 36499263 PMCID: PMC9736988 DOI: 10.3390/ijms232314940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 12/02/2022] Open
Abstract
Citrullinemia type I (CTLN1) is a rare autosomal recessive disorder caused by mutations in the gene encoding argininosuccinate synthetase 1 (ASS1) that catalyzes the third step of the urea cycle. CTLN1 patients suffer from impaired elimination of nitrogen, which leads to neurotoxic levels of circulating ammonia and urea cycle byproducts that may cause severe metabolic encephalopathy, death or irreversible brain damage. Standard of care (SOC) of CTLN1 consists of daily nitrogen-scavenger administration, but patients remain at risk of life-threatening decompensations. We evaluated the therapeutic efficacy of a recombinant adeno-associated viral vector carrying the ASS1 gene under the control of a liver-specific promoter (VTX-804). When administered to three-week-old CTLN1 mice, all the animals receiving VTX-804 in combination with SOC gained body weight normally, presented with a normalization of ammonia and reduction of citrulline levels in circulation, and 100% survived for 7 months. Similar to what has been observed in CTLN1 patients, CTLN1 mice showed several behavioral abnormalities such as anxiety, reduced welfare and impairment of innate behavior. Importantly, all clinical alterations were notably improved after treatment with VTX-804. This study demonstrates the potential of VTX-804 gene therapy for future clinical translation to CTLN1 patients.
Collapse
Affiliation(s)
- Andrea Bazo
- Division of Gene Therapy and Regulation of Gene Expression, CIMA, University of Navarra, 31008 Pamplona, Spain
| | | | - Itsaso Mauleón
- Division of Gene Therapy and Regulation of Gene Expression, CIMA, University of Navarra, 31008 Pamplona, Spain
| | - Leire Neri
- Vivet Therapeutics, S.L., 31008 Pamplona, Spain
| | - Martin Poms
- Department of Clinical Chemistry and Biochemistry, University Children’s Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Johannes Häberle
- Division of Metabolism, Children’s Research Centre (CRC), University Children’s Hospital Zurich, 8091 Zurich, Switzerland
| | - Ana Ricobaraza
- Division of Gene Therapy and Regulation of Gene Expression, CIMA, University of Navarra, 31008 Pamplona, Spain
| | | | | | - Gloria Gonzalez-Aseguinolaza
- Division of Gene Therapy and Regulation of Gene Expression, CIMA, University of Navarra, 31008 Pamplona, Spain
- Vivet Therapeutics, S.L., 31008 Pamplona, Spain
| | - Rafael Aldabe
- Division of Gene Therapy and Regulation of Gene Expression, CIMA, University of Navarra, 31008 Pamplona, Spain
| |
Collapse
|
4
|
Khoja S, Liu XB, Truong B, Nitzahn M, Lambert J, Eliav A, Nasser E, Randolph E, Burke KE, White R, Zhu X, Martini PG, Nissim I, Cederbaum SD, Lipshutz GS. Intermittent lipid nanoparticle mRNA administration prevents cortical dysmyelination associated with arginase deficiency. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 28:859-874. [PMID: 35694211 PMCID: PMC9156989 DOI: 10.1016/j.omtn.2022.04.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 04/22/2022] [Indexed: 11/28/2022]
Abstract
Arginase deficiency is associated with prominent neuromotor features, including spastic diplegia, clonus, and hyperreflexia; intellectual disability and progressive neurological decline are other signs. In a constitutive murine model, we recently described leukodystrophy as a significant component of the central nervous system features of arginase deficiency. In the present studies, we sought to examine if the administration of a lipid nanoparticle carrying human ARG1 mRNA to constitutive knockout mice could prevent abnormalities in myelination associated with arginase deficiency. Imaging of the cingulum, striatum, and cervical segments of the corticospinal tract revealed a drastic reduction of myelinated axons; signs of degenerating axons were also present with thin myelin layers. Lipid nanoparticle/ARG1 mRNA administration resulted in both light and electron microscopic evidence of a dramatic recovery of myelin density compared with age-matched controls; oligodendrocytes were seen to be extending processes to wrap many axons. Abnormally thin myelin layers, when myelination was present, were resolved with intermittent mRNA administration, indicative of not only a greater density of myelinated axons but also an increase in the thickness of the myelin sheath. In conclusion, lipid nanoparticle/ARG1 mRNA administration in arginase deficiency prevents the associated leukodystrophy and restores normal oligodendrocyte function.
Collapse
Affiliation(s)
- Suhail Khoja
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Xiao-Bo Liu
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Brian Truong
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Matthew Nitzahn
- Molecular Biology Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Jenna Lambert
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Adam Eliav
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Eram Nasser
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Emma Randolph
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | | | - Rebecca White
- Moderna Inc., 200 Technology Square, Cambridge, MA 02139, USA
| | - Xuling Zhu
- Moderna Inc., 200 Technology Square, Cambridge, MA 02139, USA
| | | | - Itzhak Nissim
- Division of Metabolism and Human Genetics, The Children Hospital of Philadelphia and The Department of Biochemistry and Biophysics, Perlman School of Medicine, Philadelphia, PA 19104, USA
| | - Stephen D. Cederbaum
- Department of Psychiatry, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Intellectual and Developmental Disabilities Research Center at UCLA, Los Angeles, CA 90095, USA
- Semel Institute for Neuroscience, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Gerald S. Lipshutz
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Molecular Biology Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Department of Psychiatry, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Intellectual and Developmental Disabilities Research Center at UCLA, Los Angeles, CA 90095, USA
- Semel Institute for Neuroscience, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
5
|
Khoja S, Lambert J, Nitzahn M, Eliav A, Zhang Y, Tamboline M, Le CT, Nasser E, Li Y, Patel P, Zhuravka I, Lueptow LM, Tkachyova I, Xu S, Nissim I, Schulze A, Lipshutz GS. Gene therapy for guanidinoacetate methyltransferase deficiency restores cerebral and myocardial creatine while resolving behavioral abnormalities. Mol Ther Methods Clin Dev 2022; 25:278-296. [PMID: 35505663 PMCID: PMC9051621 DOI: 10.1016/j.omtm.2022.03.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/27/2022] [Indexed: 11/06/2022]
Abstract
Creatine deficiency disorders are inborn errors of creatine metabolism, an energy homeostasis molecule. One of these, guanidinoacetate N-methyltransferase (GAMT) deficiency, has clinical characteristics that include features of autism, self-mutilation, intellectual disability, and seizures, with approximately 40% having a disorder of movement; failure to thrive can also be a component. Along with low creatine levels, guanidinoacetic acid (GAA) toxicity has been implicated in the pathophysiology of the disorder. Present-day therapy with oral creatine to control GAA lacks efficacy; seizures can persist. Dietary management and pharmacological ornithine treatment are challenging. Using an AAV-based gene therapy approach to express human codon-optimized GAMT in hepatocytes, in situ hybridization, and immunostaining, we demonstrated pan-hepatic GAMT expression. Serial collection of blood demonstrated a marked early and sustained reduction of GAA with normalization of plasma creatine; urinary GAA levels also markedly declined. The terminal time point demonstrated marked improvement in cerebral and myocardial creatine levels. In conjunction with the biochemical findings, treated mice gained weight to nearly match their wild-type littermates, while behavioral studies demonstrated resolution of abnormalities; PET-CT imaging demonstrated improvement in brain metabolism. In conclusion, a gene therapy approach can result in long-term normalization of GAA with increased creatine in guanidinoacetate N-methyltransferase deficiency and at the same time resolves the behavioral phenotype in a murine model of the disorder. These findings have important implications for the development of a new therapy for this abnormality of creatine metabolism.
Collapse
Affiliation(s)
- Suhail Khoja
- Department of Surgery, UCLA, Los Angeles, CA 90025, USA
| | - Jenna Lambert
- Department of Surgery, UCLA, Los Angeles, CA 90025, USA
| | - Matthew Nitzahn
- Molecular Biology Institute, UCLA, Los Angeles, CA 90025, USA
| | - Adam Eliav
- Department of Surgery, UCLA, Los Angeles, CA 90025, USA
| | - YuChen Zhang
- Semel Institute for Neuroscience, UCLA, Los Angeles, CA 90025, USA
| | - Mikayla Tamboline
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, CA 90025, USA.,Departments of Molecular and Medical Pharmacology, Universtiy of California, Los Angeles, CA 90025, USA
| | - Colleen T Le
- Department of Surgery, UCLA, Los Angeles, CA 90025, USA
| | - Eram Nasser
- Department of Surgery, UCLA, Los Angeles, CA 90025, USA
| | - Yunfeng Li
- Departments of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA 90025, USA
| | - Puja Patel
- Department of Surgery, UCLA, Los Angeles, CA 90025, USA
| | - Irina Zhuravka
- Behavioral Testing Core, Department of Psychology, UCLA, Los Angeles, CA 90025, USA
| | - Lindsay M Lueptow
- Behavioral Testing Core, Department of Psychology, UCLA, Los Angeles, CA 90025, USA
| | - Ilona Tkachyova
- Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Shili Xu
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, CA 90025, USA.,Departments of Molecular and Medical Pharmacology, Universtiy of California, Los Angeles, CA 90025, USA.,Jonsson Comprehensive Cancer Center at UCLA, David Geffen School of Medicine at UCLA, Los Angeles, CA 90025, USA
| | - Itzhak Nissim
- Division of Metabolism and Human Genetics, Children's Hospital of Philadelphia, and the Department of Biochemistry and Biophysics, Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andreas Schulze
- Department of Paediatrics, University of Toronto, Toronto, ON M5G 1X8, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON M5G 1X8, Canada.,Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Gerald S Lipshutz
- Department of Surgery, UCLA, Los Angeles, CA 90025, USA.,Molecular Biology Institute, UCLA, Los Angeles, CA 90025, USA.,Semel Institute for Neuroscience, UCLA, Los Angeles, CA 90025, USA.,Departments of Molecular and Medical Pharmacology, Universtiy of California, Los Angeles, CA 90025, USA.,Intellectual and Developmental Disabilities Research Center, UCLA, Los Angeles, CA 90025, USA
| |
Collapse
|
6
|
Córdoba KM, Jericó D, Sampedro A, Jiang L, Iraburu MJ, Martini PGV, Berraondo P, Avila MA, Fontanellas A. Messenger RNA as a personalized therapy: The moment of truth for rare metabolic diseases. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 372:55-96. [PMID: 36064267 DOI: 10.1016/bs.ircmb.2022.03.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Inborn errors of metabolism (IEM) encompass a group of monogenic diseases affecting both pediatric and adult populations and currently lack effective treatments. Some IEM such as familial hypercholesterolemia or X-linked protoporphyria are caused by gain of function mutations, while others are characterized by an impaired protein function, causing a metabolic pathway blockage. Pathophysiology classification includes intoxication, storage and energy-related metabolic disorders. Factors specific to each disease trigger acute metabolic decompensations. IEM require prompt and effective care, since therapeutic delay has been associated with the development of fatal events including severe metabolic acidosis, hyperammonemia, cerebral edema, and death. Rapid expression of therapeutic proteins can be achieved hours after the administration of messenger RNAs (mRNA), representing an etiological solution for acute decompensations. mRNA-based therapy relies on modified RNAs with enhanced stability and translatability into therapeutic proteins. The proteins produced in the ribosomes can be targeted to specific intracellular compartments, the cell membrane, or be secreted. Non-immunogenic lipid nanoparticle formulations have been optimized to prevent RNA degradation and to allow safe repetitive administrations depending on the disease physiopathology and clinical status of the patients, thus, mRNA could be also an effective chronic treatment for IEM. Given that the liver plays a key role in most of metabolic pathways or can be used as bioreactor for excretable proteins, this review focuses on the preclinical and clinical evidence that supports the implementation of mRNA technology as a promising personalized strategy for liver metabolic disorders such as acute intermittent porphyria, ornithine transcarbamylase deficiency or glycogen storage disease.
Collapse
Affiliation(s)
- Karol M Córdoba
- Hepatology Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Daniel Jericó
- Hepatology Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Ana Sampedro
- Hepatology Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Lei Jiang
- Moderna Inc, Cambridge, MA, United States
| | - María J Iraburu
- Department of Biochemistry and Genetics. School of Sciences, University of Navarra, Pamplona, Spain
| | | | - Pedro Berraondo
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain; Program of Immunology and Immunotherapy, CIMA-University of Navarra, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Matías A Avila
- Hepatology Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Fontanellas
- Hepatology Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
7
|
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: 271] [Impact Index Per Article: 45.2] [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
|
8
|
Truong B, Allegri G, Liu XB, Burke KE, Zhu X, Cederbaum SD, Häberle J, Martini PGV, Lipshutz GS. Lipid nanoparticle-targeted mRNA therapy as a treatment for the inherited metabolic liver disorder arginase deficiency. Proc Natl Acad Sci U S A 2019; 116:21150-21159. [PMID: 31501335 PMCID: PMC6800360 DOI: 10.1073/pnas.1906182116] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Arginase deficiency is caused by biallelic mutations in arginase 1 (ARG1), the final step of the urea cycle, and results biochemically in hyperargininemia and the presence of guanidino compounds, while it is clinically notable for developmental delays, spastic diplegia, psychomotor function loss, and (uncommonly) death. There is currently no completely effective medical treatment available. While preclinical strategies have been demonstrated, disadvantages with viral-based episomal-expressing gene therapy vectors include the risk of insertional mutagenesis and limited efficacy due to hepatocellular division. Recent advances in messenger RNA (mRNA) codon optimization, synthesis, and encapsulation within biodegradable liver-targeted lipid nanoparticles (LNPs) have potentially enabled a new generation of safer, albeit temporary, treatments to restore liver metabolic function in patients with urea cycle disorders, including ARG1 deficiency. In this study, we applied such technologies to successfully treat an ARG1-deficient murine model. Mice were administered LNPs encapsulating human codon-optimized ARG1 mRNA every 3 d. Mice demonstrated 100% survival with no signs of hyperammonemia or weight loss to beyond 11 wk, compared with controls that perished by day 22. Plasma ammonia, arginine, and glutamine demonstrated good control without elevation of guanidinoacetic acid, a guanidino compound. Evidence of urea cycle activity restoration was demonstrated by the ability to fully metabolize an ammonium challenge and by achieving near-normal ureagenesis; liver arginase activity achieved 54% of wild type. Biochemical and microscopic data showed no evidence of hepatotoxicity. These results suggest that delivery of ARG1 mRNA by liver-targeted nanoparticles may be a viable gene-based therapeutic for the treatment of arginase deficiency.
Collapse
Affiliation(s)
- Brian Truong
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095
| | - Gabriella Allegri
- Division of Metabolism and Children's Research Center, University Children's Hospital, 8032 Zurich, Switzerland
| | - Xiao-Bo Liu
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095
| | | | | | - Stephen D Cederbaum
- Department of Psychiatry, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095
- Intellectual and Developmental Disabilities Research Center at UCLA, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095
- Semel Institute for Neuroscience, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095
| | - Johannes Häberle
- Division of Metabolism and Children's Research Center, University Children's Hospital, 8032 Zurich, Switzerland
| | | | - Gerald S Lipshutz
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095;
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095
- Department of Psychiatry, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095
- Intellectual and Developmental Disabilities Research Center at UCLA, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095
- Semel Institute for Neuroscience, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095
| |
Collapse
|
9
|
Liu XB, Haney JR, Cantero G, Lambert JR, Otero-Garcia M, Truong B, Gropman A, Cobos I, Cederbaum SD, Lipshutz GS. Hepatic arginase deficiency fosters dysmyelination during postnatal CNS development. JCI Insight 2019; 4:130260. [PMID: 31484827 DOI: 10.1172/jci.insight.130260] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/01/2019] [Indexed: 12/26/2022] Open
Abstract
Deficiency of arginase is associated with hyperargininemia, and prominent features include spastic diplegia/tetraplegia, clonus, and hyperreflexia; loss of ambulation, intellectual disability and progressive neurological decline are other signs. To gain greater insight into the unique neuromotor features, we performed gene expression profiling of the motor cortex of a murine model of the disorder. Coexpression network analysis suggested an abnormality with myelination, which was supported by limited existing human data. Utilizing electron microscopy, marked dysmyelination was detected in 2-week-old homozygous Arg1-KO mice. The corticospinal tract was found to be adversely affected, supporting dysmyelination as the cause of the unique neuromotor features and implicating oligodendrocyte impairment in a deficiency of hepatic Arg1. Following neonatal hepatic gene therapy to express Arg1, the subcortical white matter, pyramidal tract, and corticospinal tract all showed a remarkable recovery in terms of myelinated axon density and ultrastructural integrity with active wrapping of axons by nearby oligodendrocyte processes. These findings support the following conclusions: arginase deficiency is a leukodystrophy affecting the brain and spinal cord while sparing the peripheral nervous system, and neonatal AAV hepatic gene therapy can rescue the defects associated with myelinated axons, strongly implicating the functional recovery of oligodendrocytes after restoration of hepatic arginase activity.
Collapse
Affiliation(s)
| | - Jillian R Haney
- Department of Psychiatry.,Intellectual and Developmental Disabilities Research Center, and.,Semel Institute for Neuroscience, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Gloria Cantero
- Neuromuscular Disorders Unit, Department of Neurology, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | | | - Brian Truong
- Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Andrea Gropman
- Neurogenetics and Neurodevelopmental Pediatrics and Genetics, Children's National Health System, Washington, DC, USA
| | - Inma Cobos
- Department of Pathology and Laboratory Medicine and
| | - Stephen D Cederbaum
- Department of Psychiatry.,Intellectual and Developmental Disabilities Research Center, and.,Semel Institute for Neuroscience, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Gerald S Lipshutz
- Department of Surgery.,Department of Psychiatry.,Intellectual and Developmental Disabilities Research Center, and.,Semel Institute for Neuroscience, David Geffen School of Medicine at UCLA, Los Angeles, California.,Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Molecular Biology Institute, UCLA, Los Angeles, California, USA
| |
Collapse
|
10
|
Krystofova J, Pathipati P, Russ J, Sheldon A, Ferriero D. The Arginase Pathway in Neonatal Brain Hypoxia-Ischemia. Dev Neurosci 2019; 40:437-450. [PMID: 30995639 PMCID: PMC6784534 DOI: 10.1159/000496467] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/20/2018] [Indexed: 12/11/2022] Open
Abstract
Brain damage after hypoxia-ischemia (HI) occurs in an age-dependent manner. Neuroprotective strategies assumed to be effective in adults might have deleterious effects in the immature brain. In order to create effective therapies, the complex pathophysiology of HI in the developing brain requires exploring new mechanisms. Critical determinants of neuronal survival after HI are the extent of vascular dysfunction, inflammation, and oxidative stress, followed later by tissue repair. The key enzyme of these processes in the human body is arginase (ARG) that acts via the bioavailability of nitric oxide, and the synthesis of polyamines and proline. ARG is expressed throughout the brain in different cells. However, little is known about the effect of ARG in pathophysiological states of the brain, especially hypoxia-ischemia. Here, we summarize the role of ARG during neurodevelopment as well as in various brain pathologies.
Collapse
Affiliation(s)
- Jana Krystofova
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA,
| | - Praneeti Pathipati
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Jeffrey Russ
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Ann Sheldon
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Donna Ferriero
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
11
|
Ashley SN, Nordin JML, Buza EL, Greig JA, Wilson JM. Adeno-associated viral gene therapy corrects a mouse model of argininosuccinic aciduria. Mol Genet Metab 2018; 125:241-250. [PMID: 30253962 DOI: 10.1016/j.ymgme.2018.08.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 08/26/2018] [Accepted: 08/27/2018] [Indexed: 12/11/2022]
Abstract
Argininosuccinic aciduria (ASA) is the second most common genetic disorder affecting the urea cycle. The disease is caused by deleterious mutations in the gene encoding argininosuccinate lyase (ASL); total loss of ASL activity results in severe neonatal onset of the disease, which is characterized by hyperammonemia within a few days of birth that can rapidly progress to coma and death. The long-term complications of ASA, such as hypertension and neurocognitive deficits, appear to be resistant to the current treatment options of dietary restriction, arginine supplementation, and nitrogen scavenging drugs. Treatment-resistant disease is currently being managed by orthotopic liver transplant, which shows variable improvement and requires lifetime immunosuppression. Here, we developed a gene therapy strategy for ASA aimed at alleviating the symptoms associated with urea cycle disruption by providing stable expression of ASL protein in the liver. We designed a codon-optimized human ASL gene packaged within adeno-associated virus serotype 8 (AAV8) as a vector for targeted delivery to the liver. To evaluate the therapeutic efficacy of this approach, we utilized a murine hypomorphic model of ASA. Neonatal administration of AAV8 via the temporal facial vein extended survival in ASA hypomorphic mice, although not to wild-type levels. Intravenous injection into adolescent hypomorphic mice led to increased survival and body weight and correction of metabolites associated with the disease. Our results demonstrate that AAV8 gene therapy is a viable approach for the treatment of ASA.
Collapse
Affiliation(s)
- Scott N Ashley
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jayme M L Nordin
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth L Buza
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jenny A Greig
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James M Wilson
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Attenuation of the Niemann-Pick type C2 disease phenotype by intracisternal administration of an AAVrh.10 vector expressing Npc2. Exp Neurol 2018; 306:22-33. [DOI: 10.1016/j.expneurol.2018.04.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 02/28/2018] [Accepted: 04/01/2018] [Indexed: 11/18/2022]
|
13
|
Qi F, Zuo Z, Hu S, Xia Y, Song D, Kong J, Yang Y, Wu Y, Wang X, Yang J, Hu D, Yuan Q, Zou J, Guo K, Xu J, Yao Z. An enriched environment restores hepatitis B vaccination-mediated impairments in synaptic function through IFN-γ/Arginase1 signaling. Brain Behav Immun 2018; 71:116-132. [PMID: 29627530 DOI: 10.1016/j.bbi.2018.04.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/06/2018] [Accepted: 04/04/2018] [Indexed: 12/28/2022] Open
Abstract
Activation of the neonatal immune system may contribute to deficits in neuronal plasticity. We have reported that neonatal vaccination with a hepatitis B vaccine (HBV) transiently impairs mood status and spatial memory involving a systemic T helper (Th) 2 bias and M1 microglial activation. Here, an EE induced microglial anti-inflammatory M2 polarization, as evidenced by selectively enhanced expression of the Arginase1 gene (Arg-1) in the hippocampus. Interestingly, knock-down of the Arg-1 gene prevented the effects of EE on restoring the dendritic spine density. Moreover, levels of the Th1-derived cytokine IFN-gamma (IFN-γ) were elevated in the choroid plexus (CP), which is the interface between the brain and the periphery. IFN-γ-blocking antibodies blunted the protective effects of an EE on spine density and LTP. Furthermore, levels of complement proteins C1q and C3 were elevated, and this elevation was associated with synapse loss induced by the HBV, whereas an EE reversed the effects of the HBV. Similarly, blockade of C1q activation clearly prevented synaptic pruning by microglia, LTP inhibition and memory deficits in hepatitis B-vaccinated mice. Together, the EE-induced increase in IFN-γ levels in the CP may disrupt systemic immunosuppression related to HBV via an IFN-γ/Arg-1/complement-dependent pathway.
Collapse
Affiliation(s)
- Fangfang Qi
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Zejie Zuo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Saisai Hu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Yucen Xia
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, 232 Waihuandong Road, Guangzhou 510006, PR China
| | - Dan Song
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Jiechen Kong
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Yang Yang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Yingying Wu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Xiao Wang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Junhua Yang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Dandan Hu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Department of Neurology, Guangzhou Women and Children's Hospital, #9, Jinsui Road, Guangzhou 510623, PR China
| | - Qunfang Yuan
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Juntao Zou
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Kaihua Guo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Jie Xu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Zhibin Yao
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China.
| |
Collapse
|
14
|
Diez-Fernandez C, Rüfenacht V, Gemperle C, Fingerhut R, Häberle J. Mutations and common variants in the human arginase 1 (ARG1
) gene: Impact on patients, diagnostics, and protein structure considerations. Hum Mutat 2018; 39:1029-1050. [DOI: 10.1002/humu.23545] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/20/2018] [Accepted: 04/25/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Carmen Diez-Fernandez
- University Children's Hospital Zurich; Division of Metabolism and Children's Research Center; Zurich Switzerland
| | - Véronique Rüfenacht
- University Children's Hospital Zurich; Division of Metabolism and Children's Research Center; Zurich Switzerland
| | - Corinne Gemperle
- University Children's Hospital Zurich; Division of Metabolism and Children's Research Center; Zurich Switzerland
| | - Ralph Fingerhut
- University Children's Hospital Zurich; Division of Metabolism and Children's Research Center; Zurich Switzerland
| | - Johannes Häberle
- University Children's Hospital Zurich; Division of Metabolism and Children's Research Center; Zurich Switzerland
| |
Collapse
|
15
|
Angarita SAK, Truong B, Khoja S, Nitzahn M, Rajbhandari AK, Zhuravka I, Duarte S, Lin MG, Lam AK, Cederbaum SD, Lipshutz GS. Human hepatocyte transplantation corrects the inherited metabolic liver disorder arginase deficiency in mice. Mol Genet Metab 2018; 124:114-123. [PMID: 29724658 PMCID: PMC5976549 DOI: 10.1016/j.ymgme.2018.04.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/16/2018] [Accepted: 04/16/2018] [Indexed: 12/13/2022]
Abstract
The transplantation, engraftment, and expansion of primary hepatocytes have the potential to be an effective therapy for metabolic disorders of the liver including those of nitrogen metabolism. To date, such methods for the treatment of urea cycle disorders in murine models has only been minimally explored. Arginase deficiency, an inherited disorder of nitrogen metabolism that presents in the first two years of life, has the potential to be treated by such methods. To explore the potential of this approach, we mated the conditional arginase deficient mouse with a mouse model deficient in fumarylacetoacetate hydrolase (FAH) and with Rag2 and IL2-Rγ mutations to give a selective advantage to transplanted (normal) human hepatocytes. On day -1, a uroplasminogen-expressing adenoviral vector was administered intravenously followed the next day with the transplantation of 1 × 106 human hepatocytes (or vehicle alone) by intrasplenic injection. As the initial number of administered hepatocytes would be too low to prevent hepatotoxicity-induced mortality, NTBC cycling was performed to allow for hepatocyte expansion and repopulation. While all control mice died, all except one human hepatocyte transplanted mice survived. Four months after hepatocyte transplantation, 2 × 1011 genome copies of AAV-TBG-Cre recombinase was administered IV to disrupt endogenous hepatic arginase expression. While all control mice died within the first month, human hepatocyte transplanted mice did well. Ammonia and amino acids, analyzed in both groups before and after disruption of endogenous arginase expression, while well-controlled in the transplanted group, were markedly abnormal in the controls. Ammonium challenging further demonstrated the durability and functionality of the human repopulated liver. In conclusion, these studies demonstrate that human hepatocyte repopulation in the murine liver can result in effective treatment of arginase deficiency.
Collapse
Affiliation(s)
- Stephanie A K Angarita
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Brian Truong
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Suhail Khoja
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Matthew Nitzahn
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States; Molecular Biology Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Abha K Rajbhandari
- Behavioral Testing Core Facility, Department of Psychology and Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Irina Zhuravka
- Behavioral Testing Core Facility, Department of Psychology and Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Sergio Duarte
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Michael G Lin
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Alex K Lam
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Stephen D Cederbaum
- Department of Psychiatry, The David Geffen School of Medicine at UCLA, Los Angeles, CA, United States; Intellectual and Developmental Disabilities Research Center at UCLA, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Gerald S Lipshutz
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States; Molecular Biology Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States; Department of Psychiatry, The David Geffen School of Medicine at UCLA, Los Angeles, CA, United States; Intellectual and Developmental Disabilities Research Center at UCLA, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States; Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States; Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States; Semel Institute for Neuroscience, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States; Broad Center for Regenerative Medicine and Stem Cell Research at UCLA, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States.
| |
Collapse
|
16
|
Rescue of the Functional Alterations of Motor Cortical Circuits in Arginase Deficiency by Neonatal Gene Therapy. J Neurosci 2017; 36:6680-90. [PMID: 27335400 DOI: 10.1523/jneurosci.0897-16.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 05/11/2016] [Indexed: 01/28/2023] Open
Abstract
UNLABELLED Arginase 1 deficiency is a urea cycle disorder associated with hyperargininemia, spastic diplegia, loss of ambulation, intellectual disability, and seizures. To gain insight on how loss of arginase expression affects the excitability and synaptic connectivity of the cortical neurons in the developing brain, we used anatomical, ultrastructural, and electrophysiological techniques to determine how single-copy and double-copy arginase deletion affects cortical circuits in mice. We find that the loss of arginase 1 expression results in decreased dendritic complexity, decreased excitatory and inhibitory synapse numbers, decreased intrinsic excitability, and altered synaptic transmission in layer 5 motor cortical neurons. Hepatic arginase 1 gene therapy using adeno-associated virus rescued nearly all these abnormalities when administered to neonatal homozygous knock-out animals. Therefore, gene therapeutic strategies can reverse physiological and anatomical markers of arginase 1 deficiency and therefore may be of therapeutic benefit for the neurological disabilities in this syndrome. SIGNIFICANCE STATEMENT These studies are one of the few investigations to try to understand the underlying neurological dysfunction that occurs in urea cycle disorders and the only to examine arginase deficiency. We have demonstrated by multiple modalities that, in murine layer 5 cortical neurons, a gradation of abnormalities exists based on the functional copy number of arginase: intrinsic excitability is altered, there is decreased density in asymmetrical and perisomatic synapses, and analysis of the dendritic complexity is lowest in the homozygous knock-out. With neonatal administration of adeno-associated virus expressing arginase, there is near-total recovery of the abnormalities in neurons and cortical circuits, supporting the concept that neonatal gene therapy may prevent the functional abnormalities that occur in arginase deficiency.
Collapse
|
17
|
Sin YY, Price PR, Ballantyne LL, Funk CD. Proof-of-Concept Gene Editing for the Murine Model of Inducible Arginase-1 Deficiency. Sci Rep 2017; 7:2585. [PMID: 28566761 PMCID: PMC5451454 DOI: 10.1038/s41598-017-02927-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 04/28/2017] [Indexed: 12/14/2022] Open
Abstract
Arginase-1 deficiency in humans is a rare genetic disorder of metabolism resulting from a loss of arginase-1, leading to impaired ureagenesis, hyperargininemia and neurological deficits. Previously, we generated a tamoxifen-inducible arginase-1 deficient mouse model harboring a deletion of Arg1 exons 7 and 8 that leads to similar biochemical defects, along with a wasting phenotype and death within two weeks. Here, we report a strategy utilizing the Clustered, Regularly Interspaced, Short Palindromic Repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system in conjunction with piggyBac technology to target and reincorporate exons 7 and 8 at the specific Arg1 locus in attempts to restore the function of arginase-1 in induced pluripotent stem cell (iPSC)-derived hepatocyte-like cells (iHLCs) and macrophages in vitro. While successful gene targeted repair was achieved, minimal urea cycle function was observed in the targeted iHLCs compared to adult hepatocytes likely due to inadequate maturation of the cells. On the other hand, iPSC-derived macrophages expressed substantial amounts of "repaired" arginase. Our studies provide proof-of-concept for gene-editing at the Arg1 locus and highlight the challenges that lie ahead to restore sufficient liver-based urea cycle function in patients with urea cycle disorders.
Collapse
Affiliation(s)
- Yuan Yan Sin
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Phillipe R Price
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Laurel L Ballantyne
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Colin D Funk
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
18
|
Lee PC, Truong B, Vega-Crespo A, Gilmore WB, Hermann K, Angarita SA, Tang JK, Chang KM, Wininger AE, Lam AK, Schoenberg BE, Cederbaum SD, Pyle AD, Byrne JA, Lipshutz GS. Restoring Ureagenesis in Hepatocytes by CRISPR/Cas9-mediated Genomic Addition to Arginase-deficient Induced Pluripotent Stem Cells. MOLECULAR THERAPY-NUCLEIC ACIDS 2016; 5:e394. [PMID: 27898091 PMCID: PMC5155330 DOI: 10.1038/mtna.2016.98] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/26/2016] [Indexed: 12/18/2022]
Abstract
Urea cycle disorders are incurable enzymopathies that affect nitrogen metabolism and typically lead to hyperammonemia. Arginase deficiency results from a mutation in Arg1, the enzyme regulating the final step of ureagenesis and typically results in developmental disabilities, seizures, spastic diplegia, and sometimes death. Current medical treatments for urea cycle disorders are only marginally effective, and for proximal disorders, liver transplantation is effective but limited by graft availability. Advances in human induced pluripotent stem cell research has allowed for the genetic modification of stem cells for potential cellular replacement therapies. In this study, we demonstrate a universally-applicable CRISPR/Cas9-based strategy utilizing exon 1 of the hypoxanthine-guanine phosphoribosyltransferase locus to genetically modify and restore arginase activity, and thus ureagenesis, in genetically distinct patient-specific human induced pluripotent stem cells and hepatocyte-like derivatives. Successful strategies restoring gene function in patient-specific human induced pluripotent stem cells may advance applications of genetically modified cell therapy to treat urea cycle and other inborn errors of metabolism.
Collapse
Affiliation(s)
- Patrick C Lee
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Brian Truong
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Agustin Vega-Crespo
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - W Blake Gilmore
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Kip Hermann
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Stephanie Ak Angarita
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Jonathan K Tang
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Katherine M Chang
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Austin E Wininger
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Alex K Lam
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Benjamen E Schoenberg
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Stephen D Cederbaum
- Department of Psychiatry, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Intellectual and Developmental Disabilities Research Center at UCLA, Los Angeles, California, USA.,Semel Institute for Neuroscience, UCLA, Los Angeles, California, USA
| | - April D Pyle
- Department of Microbiology, Immunology and Molecular Genetics, UCLA, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - James A Byrne
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Gerald S Lipshutz
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Department of Psychiatry, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Intellectual and Developmental Disabilities Research Center at UCLA, Los Angeles, California, USA.,Semel Institute for Neuroscience, UCLA, Los Angeles, California, USA.,Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
19
|
A Novel Pathway for Metabolism of the Cardiovascular Risk Factor Homoarginine by alanine:glyoxylate aminotransferase 2. Sci Rep 2016; 6:35277. [PMID: 27752063 PMCID: PMC5082758 DOI: 10.1038/srep35277] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 09/23/2016] [Indexed: 01/06/2023] Open
Abstract
Low plasma concentrations of L-homoarginine are associated with an increased risk of cardiovascular events, while homoarginine supplementation is protective in animal models of metabolic syndrome and stroke. Catabolism of homoarginine is still poorly understood. Based on the recent findings from a Genome Wide Association Study we hypothesized that homoarginine can be metabolized by alanine:glyoxylate aminotransferase 2 (AGXT2). We purified human AGXT2 from tissues of AGXT2 transgenic mice and demonstrated its ability to metabolize homoarginine to 6-guanidino-2-oxocaproic acid (GOCA). After incubation of HepG2 cells overexpressing AGXT2 with isotope-labeled homoarginine-d4 we were able to detect labeled GOCA in the medium. We injected wild type mice with labeled homoarginine and detected labeled GOCA in the plasma. We found that AGXT2 knockout (KO) mice have higher homoarginine and lower GOCA plasma levels as compared to wild type mice, while the reverse was true for AGXT2 transgenic (Tg) mice. In summary, we experimentally proved the presence of a new pathway of homoarginine catabolism – its transamination by AGXT2 with formation of GOCA and demonstrated that endogenous AGXT2 is required for maintenance of homoarginine levels in mice. Our findings may lead to development of novel therapeutic approaches for cardiovascular pathologies associated with homoarginine deficiency.
Collapse
|
20
|
Liver-specific knockout of arginase-1 leads to a profound phenotype similar to inducible whole body arginase-1 deficiency. Mol Genet Metab Rep 2016; 9:54-60. [PMID: 27761413 PMCID: PMC5065044 DOI: 10.1016/j.ymgmr.2016.10.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 10/05/2016] [Indexed: 12/27/2022] Open
Abstract
Arginase-1 (Arg1) converts arginine to urea and ornithine in the distal step of the urea cycle in liver. We previously generated a tamoxifen-inducible Arg1 deficient mouse model (Arg1-Cre) that disrupts Arg1 expression throughout the whole body and leads to lethality ≈ 2 weeks after gene disruption. Here, we evaluate if liver-selective Arg1 loss is sufficient to recapitulate the phenotype observed in global Arg1 knockout mice, as well as to gauge the effectiveness of gene delivery or hepatocyte transplantation to rescue the phenotype. Liver-selective Arg1 deletion was induced by using an adeno-associated viral (AAV)-thyroxine binding globulin (TBG) promoter-Cre recombinase vector administered to Arg1 “floxed” mice; Arg1fl/fl). An AAV vector expressing an Arg1-enhanced green fluorescent protein (Arg1-eGFP) transgene was used for gene delivery, while intrasplenic injection of wild-type (WT) C57BL/6 hepatocytes after partial hepatectomy was used for cell delivery to “rescue” tamoxifen-treated Arg1-Cre mice. The results indicate that liver-selective loss of Arg1 (> 90% deficient) leads to a phenotype resembling the whole body knockout of Arg1 with lethality ≈ 3 weeks after Cre-induced gene disruption. Delivery of Arg1-eGFP AAV rescues more than half of Arg1 global knockout male mice (survival > 4 months) but a significant proportion still succumb to the enzyme deficiency even though liver expression and enzyme activity of the fusion protein reach levels observed in WT animals. Significant Arg1 enzyme activity from engrafted WT hepatocytes into knockout livers can be achieved but not sufficient for rescuing the lethal phenotype. This raises a conundrum relating to liver-specific expression of Arg1. On the one hand, loss of expression in this organ appears to be both necessary and sufficient to explain the lethal phenotype of the genetic disorder in mice. On the other hand, gene and cell-directed therapies suggest that rescue of extra-hepatic Arg1 expression may also be necessary for disease correction. Further studies are needed in order to illuminate the detailed mechanisms for pathogenesis of Arg1-deficiency.
Collapse
|
21
|
Sin YY, Baron G, Schulze A, Funk CD. Arginase-1 deficiency. J Mol Med (Berl) 2015; 93:1287-96. [PMID: 26467175 DOI: 10.1007/s00109-015-1354-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 09/14/2015] [Accepted: 10/01/2015] [Indexed: 12/13/2022]
Abstract
Arginase-1 (ARG1) deficiency is a rare autosomal recessive disorder that affects the liver-based urea cycle, leading to impaired ureagenesis. This genetic disorder is caused by 40+ mutations found fairly uniformly spread throughout the ARG1 gene, resulting in partial or complete loss of enzyme function, which catalyzes the hydrolysis of arginine to ornithine and urea. ARG1-deficient patients exhibit hyperargininemia with spastic paraparesis, progressive neurological and intellectual impairment, persistent growth retardation, and infrequent episodes of hyperammonemia, a clinical pattern that differs strikingly from other urea cycle disorders. This review briefly highlights the current understanding of the etiology and pathophysiology of ARG1 deficiency derived from clinical case reports and therapeutic strategies stretching over several decades and reports on several exciting new developments regarding the pathophysiology of the disorder using ARG1 global and inducible knockout mouse models. Gene transfer studies in these mice are revealing potential therapeutic options that can be exploited in the future. However, caution is advised in extrapolating results since the lethal disease phenotype in mice is much more severe than in humans indicating that the mouse models may not precisely recapitulate human disease etiology. Finally, some of the functions and implications of ARG1 in non-urea cycle activities are considered. Lingering questions and future areas to be addressed relating to the clinical manifestations of ARG1 deficiency in liver and brain are also presented. Hopefully, this review will spark invigorated research efforts that lead to treatments with better clinical outcomes.
Collapse
Affiliation(s)
- Yuan Yan Sin
- Department of Biomedical and Molecular Sciences, Queen's University, 433 Botterell Hall, 18 Stuart Street, Kingston, ON, K7L 3N6, Canada
| | - Garrett Baron
- Department of Biomedical and Molecular Sciences, Queen's University, 433 Botterell Hall, 18 Stuart Street, Kingston, ON, K7L 3N6, Canada
| | - Andreas Schulze
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children and University of Toronto, Toronto, ON, Canada.,Genetics and Genome Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Colin D Funk
- Department of Biomedical and Molecular Sciences, Queen's University, 433 Botterell Hall, 18 Stuart Street, Kingston, ON, K7L 3N6, Canada.
| |
Collapse
|
22
|
Ballantyne LL, Sin YY, St. Amand T, Si J, Goossens S, Haenebalcke L, Haigh JJ, Kyriakopoulou L, Schulze A, Funk CD. Strategies to rescue the consequences of inducible arginase-1 deficiency in mice. PLoS One 2015; 10:e0125967. [PMID: 25938595 PMCID: PMC4418594 DOI: 10.1371/journal.pone.0125967] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Accepted: 03/27/2015] [Indexed: 12/12/2022] Open
Abstract
Arginase-1 catalyzes the conversion of arginine to ornithine and urea, which is the final step of the urea cycle used to remove excess ammonia from the body. Arginase-1 deficiency leads to hyperargininemia in mice and man with severe lethal consequences in the former and progressive neurological impairment to varying degrees in the latter. In a tamoxifen-induced arginase-1 deficient mouse model, mice succumb to the enzyme deficiency within 2 weeks after inducing the knockout and retain <2 % enzyme in the liver. Standard clinical care regimens for arginase-1 deficiency (low-protein diet, the nitrogen-scavenging drug sodium phenylbutyrate, ornithine supplementation) either failed to extend lifespan (ornithine) or only minimally prolonged lifespan (maximum 8 days with low-protein diet and drug). A conditional, tamoxifen-inducible arginase-1 transgenic mouse strain expressing the enzyme from the Rosa26 locus modestly extended lifespan of neonatal mice, but not that of 4-week old mice, when crossed to the inducible arginase-1 knockout mouse strain. Delivery of an arginase-1/enhanced green fluorescent fusion construct by adeno-associated viral delivery (rh10 serotype with a strong cytomegalovirus-chicken β-actin hybrid promoter) rescued about 30% of male mice with lifespan prolongation to at least 6 months, extensive hepatic expression and restoration of significant enzyme activity in liver. In contrast, a vector of the AAV8 serotype driven by the thyroxine-binding globulin promoter led to weaker liver expression and did not rescue arginase-1 deficient mice to any great extent. Since the induced arginase-1 deficient mouse model displays a much more severe phenotype when compared to human arginase-1 deficiency, these studies reveal that it may be feasible with gene therapy strategies to correct the various manifestations of the disorder and they provide optimism for future clinical studies.
Collapse
Affiliation(s)
- Laurel L. Ballantyne
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Yuan Yan Sin
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Tim St. Amand
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Joshua Si
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Steven Goossens
- Vascular Cell Biology Unit, VIB Inflammation Research Center, Ghent, Belgium
- Department for Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Mammalian Functional Genetics Laboratory, Division of Blood Cancers, Australian Centre for Blood Diseases, Department of Clinical Haematology, Monash University and Alfred Health Centre, Melbourne, Australia
| | - Lieven Haenebalcke
- Vascular Cell Biology Unit, VIB Inflammation Research Center, Ghent, Belgium
- Department for Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Jody J. Haigh
- Vascular Cell Biology Unit, VIB Inflammation Research Center, Ghent, Belgium
- Department for Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Mammalian Functional Genetics Laboratory, Division of Blood Cancers, Australian Centre for Blood Diseases, Department of Clinical Haematology, Monash University and Alfred Health Centre, Melbourne, Australia
| | - Lianna Kyriakopoulou
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Andreas Schulze
- Division of Clinical & Metabolic Genetics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
- Genetics and Genome Biology Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Colin D. Funk
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
- * E-mail:
| |
Collapse
|
23
|
Possible sources and functions of l-homoarginine in the brain: review of the literature and own findings. Amino Acids 2015; 47:1729-40. [DOI: 10.1007/s00726-015-1960-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 03/05/2015] [Indexed: 11/30/2022]
|
24
|
Hu C, Tai DS, Park H, Cantero-Nieto G, Chan E, Yudkoff M, Cederbaum SD, Lipshutz GS. Minimal ureagenesis is necessary for survival in the murine model of hyperargininemia treated by AAV-based gene therapy. Gene Ther 2015; 22:111-5. [PMID: 25474440 PMCID: PMC4320015 DOI: 10.1038/gt.2014.106] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 09/15/2014] [Accepted: 10/27/2014] [Indexed: 02/07/2023]
Abstract
Hyperammonemia is less severe in arginase 1 deficiency compared with other urea cycle defects. Affected patients manifest hyperargininemia and infrequent episodes of hyperammonemia. Patients typically suffer from neurological impairment with cortical and pyramidal tract deterioration, spasticity, loss of ambulation, seizures and intellectual disability; death is less common than with other urea cycle disorders. In a mouse model of arginase I deficiency, the onset of symptoms begins with weight loss and gait instability, which progresses toward development of tail tremor with seizure-like activity; death typically occurs at about 2 weeks of life. Adeno-associated viral vector gene replacement strategies result in long-term survival of mice with this disorder. With neonatal administration of vector, the viral copy number in the liver greatly declines with hepatocyte proliferation in the first 5 weeks of life. Although the animals do survive, it is not known from a functional standpoint how well the urea cycle is functioning in the adult animals that receive adeno-associated virus. In these studies, we administered [1-13C] acetate to both littermate controls and adeno-associated virus-treated arginase 1 knockout animals and examined flux through the urea cycle. Circulating ammonia levels were mildly elevated in treated animals. Arginine and glutamine also had perturbations. Assessment 30 min after acetate administration demonstrated that ureagenesis was present in the treated knockout liver at levels as low at 3.3% of control animals. These studies demonstrate that only minimal levels of hepatic arginase activity are necessary for survival and ureagenesis in arginase-deficient mice and that this level of activity results in control of circulating ammonia. These results may have implications for potential therapy in humans with arginase deficiency.
Collapse
Affiliation(s)
- Chuhong Hu
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Denise S. Tai
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Hana Park
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Gloria Cantero-Nieto
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Emily Chan
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Marc Yudkoff
- Division of Metabolic Disease, Department of Pediatrics, Children’s Hospital of Philadelphia
| | - Stephen D. Cederbaum
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California
- Department of Psychiatry, David Geffen School of Medicine at UCLA, Los Angeles, California
- Department of Intellectual and Developmental Disabilities Research Center at UCLA, David Geffen School of Medicine at UCLA, Los Angeles, California
- The Semel Institute for Neuroscience, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Gerald S. Lipshutz
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California
- Department of Psychiatry, David Geffen School of Medicine at UCLA, Los Angeles, California
- Department of Intellectual and Developmental Disabilities Research Center at UCLA, David Geffen School of Medicine at UCLA, Los Angeles, California
- The Semel Institute for Neuroscience, David Geffen School of Medicine at UCLA, Los Angeles, California
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
25
|
Torres-Vega MA, Vargas-Jerónimo RY, Montiel-Martínez AG, Muñoz-Fuentes RM, Zamorano-Carrillo A, Pastor AR, Palomares LA. Delivery of glutamine synthetase gene by baculovirus vectors: a proof of concept for the treatment of acute hyperammonemia. Gene Ther 2014; 22:58-64. [DOI: 10.1038/gt.2014.89] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 07/26/2014] [Accepted: 09/02/2014] [Indexed: 12/27/2022]
|
26
|
Hu C, Kasten J, Park H, Bhargava R, Tai DS, Grody WW, Nguyen QG, Hauschka SD, Cederbaum SD, Lipshutz GS. Myocyte-mediated arginase expression controls hyperargininemia but not hyperammonemia in arginase-deficient mice. Mol Ther 2014; 22:1792-802. [PMID: 24888478 DOI: 10.1038/mt.2014.99] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 05/18/2014] [Indexed: 12/15/2022] Open
Abstract
Human arginase deficiency is characterized by hyperargininemia and infrequent episodes of hyperammonemia that cause neurological impairment and growth retardation. We previously developed a neonatal mouse adeno-associated viral vector (AAV) rh10-mediated therapeutic approach with arginase expressed by a chicken β-actin promoter that controlled plasma ammonia and arginine, but hepatic arginase declined rapidly. This study tested a codon-optimized arginase cDNA and compared the chicken β-actin promoter to liver- and muscle-specific promoters. ARG1(-/-) mice treated with AAVrh10 carrying the liver-specific promoter also exhibited long-term survival and declining hepatic arginase accompanied by the loss of AAV episomes during subsequent liver growth. Although arginase expression in striated muscle was not expected to counteract hyperammonemia, due to muscle's lack of other urea cycle enzymes, we hypothesized that the postmitotic phenotype in muscle would allow vector genomes to persist, and hence contribute to decreased plasma arginine. As anticipated, ARG1(-/-) neonatal mice treated with AAVrh10 carrying a modified creatine kinase-based muscle-specific promoter did not survive longer than controls; however, their plasma arginine levels remained normal when animals were hyperammonemic. These data imply that plasma arginine can be controlled in arginase deficiency by muscle-specific expression, thus suggesting an alternative approach to utilizing the liver for treating hyperargininemia.
Collapse
Affiliation(s)
- Chuhong Hu
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Jennifer Kasten
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Hana Park
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Ragini Bhargava
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Denise S Tai
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Wayne W Grody
- 1] Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA [2] Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Quynh G Nguyen
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
| | - Stephen D Hauschka
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
| | - Stephen D Cederbaum
- 1] Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California, USA [2] Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California, USA [3] Department of Psychiatry, David Geffen School of Medicine at UCLA, Los Angeles, California, USA [4] The Intellectual and Developmental Disabilities Research Center at UCLA, David Geffen School of Medicine at UCLA, Los Angeles, California, USA [5] The Semel Institute for Neuroscience, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Gerald S Lipshutz
- 1] Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA [2] Department of Psychiatry, David Geffen School of Medicine at UCLA, Los Angeles, California, USA [3] The Intellectual and Developmental Disabilities Research Center at UCLA, David Geffen School of Medicine at UCLA, Los Angeles, California, USA [4] The Semel Institute for Neuroscience, David Geffen School of Medicine at UCLA, Los Angeles, California, USA [5] Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA [6] Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA [7] Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
27
|
Sin YY, Ballantyne LL, Mukherjee K, St. Amand T, Kyriakopoulou L, Schulze A, Funk CD. Inducible arginase 1 deficiency in mice leads to hyperargininemia and altered amino acid metabolism. PLoS One 2013; 8:e80001. [PMID: 24224027 PMCID: PMC3817112 DOI: 10.1371/journal.pone.0080001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 10/01/2013] [Indexed: 01/06/2023] Open
Abstract
Arginase deficiency is a rare autosomal recessive disorder resulting from a loss of the liver arginase isoform, arginase 1 (ARG1), which is the final step in the urea cycle for detoxifying ammonia. ARG1 deficiency leads to hyperargininemia, characterized by progressive neurological impairment, persistent growth retardation and infrequent episodes of hyperammonemia. Using the Cre/loxP-directed conditional gene knockout system, we generated an inducible Arg1-deficient mouse model by crossing “floxed” Arg1 mice with CreERT2 mice. The resulting mice (Arg-Cre) die about two weeks after tamoxifen administration regardless of the starting age of inducing the knockout. These treated mice were nearly devoid of Arg1 mRNA, protein and liver arginase activity, and exhibited symptoms of hyperammonemia. Plasma amino acid analysis revealed pronounced hyperargininemia and significant alterations in amino acid and guanidino compound metabolism, including increased citrulline and guanidinoacetic acid. Despite no alteration in ornithine levels, concentrations of other amino acids such as proline and the branched-chain amino acids were reduced. In summary, we have generated and characterized an inducible Arg1-deficient mouse model exhibiting several pathologic manifestations of hyperargininemia. This model should prove useful for exploring potential treatment options of ARG1 deficiency.
Collapse
Affiliation(s)
- Yuan Yan Sin
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Laurel L. Ballantyne
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Kamalika Mukherjee
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Tim St. Amand
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Lianna Kyriakopoulou
- Division of Clinical and Metabolic Genetics, and Research Institute, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Andreas Schulze
- Division of Clinical and Metabolic Genetics, and Research Institute, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Colin D. Funk
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
- * E-mail:
| |
Collapse
|
28
|
Kasten J, Hu C, Bhargava R, Park H, Tai D, Byrne JA, Marescau B, De Deyn PP, Schlichting L, Grody WW, Cederbaum SD, Lipshutz GS. Lethal phenotype in conditional late-onset arginase 1 deficiency in the mouse. Mol Genet Metab 2013; 110:222-30. [PMID: 23920045 PMCID: PMC3800271 DOI: 10.1016/j.ymgme.2013.06.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 06/25/2013] [Accepted: 06/25/2013] [Indexed: 02/03/2023]
Abstract
Human arginase deficiency is characterized by hyperargininemia and infrequent episodes of hyperammonemia, which lead to neurological impairment with spasticity, loss of ambulation, seizures, and severe mental and growth retardation; uncommonly, patients suffer early death from this disorder. In a murine targeted knockout model, onset of the phenotypic abnormality is heralded by weight loss at around day 15, and death occurs typically by postnatal day 17 with hyperargininemia and markedly elevated ammonia. This discrepancy between the more attenuated juvenile-onset human disease and the lethal neonatal murine model has remained suboptimal for studying and developing therapy for the more common presentation of arginase deficiency. These investigations aimed to address this issue by creating an adult conditional knockout mouse to determine whether later onset of arginase deficiency also resulted in lethality. Animal survival and ammonia levels, body weight, circulating amino acids, and tissue arginase levels were examined as outcome parameters after widespread Cre-recombinase activation in a conditional knockout model of arginase 1 deficiency. One hundred percent of adult female and 70% of adult male mice died an average of 21.0 and 21.6 days, respectively, after the initiation of tamoxifen administration. Animals demonstrated elevated circulating ammonia and arginine at the onset of phenotypic abnormalities. In addition, brain and liver amino acids demonstrated abnormalities. These studies demonstrate that (a) the absence of arginase in adult animals results in a disease profile (leading to death) similar to that of the targeted knockout and (b) the phenotypic abnormalities seen in the juvenile-onset model are not exclusive to the age of the animal but instead to the biochemistry of the disorder. This adult model will be useful for developing gene- and cell-based therapies for this disorder that will not be limited by the small animal size of neonatal therapy and for developing a better understanding of the characteristics of hyperargininemia.
Collapse
Affiliation(s)
- Jennifer Kasten
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Chuhong Hu
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ragini Bhargava
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Hana Park
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Denise Tai
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - James A. Byrne
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Bart Marescau
- Laboratory of Neurochemistry and Behavior, University of Antwerp and Institute Born Bunge, Antwerp, Belgium
| | - Peter P. De Deyn
- Laboratory of Neurochemistry and Behavior, University of Antwerp and Institute Born Bunge, Antwerp, Belgium
| | - Lisa Schlichting
- Biochemical Genetics Laboratory, University of Colorado, Denver, CO, USA
| | - Wayne W. Grody
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Stephen D. Cederbaum
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Department of Psychiatry, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Intellectual and Developmental Disabilities Research Center at UCLA, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Gerald S. Lipshutz
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Intellectual and Developmental Disabilities Research Center at UCLA, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Semel Institute for Neuroscience, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|